Comprehensive Bioactive Natural Products Volume 2
Efficacy, Safety & Clinical Evaluation I
V.K. GUPTA Indian Institute of Integrative Medicine (Council of Scientific & Industrial Research) Canal Road, Jammu (J&K State)- 180 001, India
2010
CID
Studium Press LLC, U.S.A.
"This page is Intentionally Left Blank"
Comprehensive Bioactive Natural Products
VoL 2: Efficacy, Safety & Clinical Evaluation I © Copyright 2010
This book contains information obtained from authentic and highly regarded sources. Reprinted material is quoted with permission, and sources are indicated. A wide variety of references are listed. Reasonable efforts have been made to publish reliable data and information, but the editor and the publisher cannot assume responsibility for the validity of all materials or for the consequences of their use. All rights are reserved under International and Pan-American Copyright Conventions. Apart from any fair dealing for the purpose of private study, research, criticism or review, as permitted under the Copyright Act, 1956, no part of this publication may be reproduced, stored in a retrieval system or transmitted, in any form or by any means-electronic, electrical, chemical, mechanical, optical, photocopying, recording or otherwise-without the prior permission of the copyright owner.
ISBN: 1-933699-52-3 SERIES ISBN: 1-933699-50-7
Published by:
STUDIUM PRESS, LLC P.O. Box-722200, Houston, Texas-77072, USA Tel. 713-541-9400; Fax:713-541-9401 E-mail:
[email protected]
Printed at:
Thomson Press (India) Ltd.
"This page is Intentionally Left Blank"
Comprehensive Bioactive Natural Products: (Multi-Volume Set) Series Editor: V.K. Gupta E-mail:
[email protected];
[email protected] Volumes Published (2010) VoL 1: Potential & Challenges Ed. V.K Gupta Vol. 2: Efficacy, Safety & Clinical Evaluation I Ed. V.K Gupta Vol. 3: Efficacy, Safety & Clinical Evaluation II Ed. V.K Gupta Vol. 4: Antioxidants & Nutraceuticals Eds. V.K Gupta & Anil K Verma Vol. 5: Immune-modulation & Vaccine Adjuvants Ed. V.K Gupta Vol. 6: Extraction, Isolation & Characterization Eds. V.K Gupta, S.C. Taneja & B.D. Gupta Vol. 7: Structural Modifications & Drug Development Eds. V.K Gupta, S.C. Taneja & B.D. Gupta Vol. 8: Quality Control & Standardization Eds. V.K. Gupta, S.C. Taneja & B.D. Gupta
MEMBERS Dr. A. Panossian: Swedish Herbal Institute Research and Development, Kovlingevagen 21, Vallberga, SE-312 50, Sweden; E-mail:
[email protected] Prof. Yu Zhao: Department ofTCM & Natural Drug Research, College of Pharmaceutical Sciences, Room 513, Zhejiang University, Zijingang Campus, 388 Yuhangtang Rd., Hangzhou 310058, China; Email: dryuzhao®Zju.edu.cn;
[email protected] Prof. A. Evidente: Department of Organic Chemistry, Dipartimento di Scienze del Suolo, della Pianta, dell'Ambiente e delle Produzioni Animali, Universita di Napoli Federico II, Portici, Italy; E-mail:
[email protected] Prof. Mirian Salvador: Instituto de Biotecnologia, Universidade de Caxias do SuI, Rua Francisco Getulio Vargas, 1130, CEP 95070-560, Caxias do SuI, Rio Grande do SuI, Brazil; E-mail:
[email protected]
Dr. Gregory Beck: Department of Biology, University of Massachusetts Boston, 100 Morrissey Blvd., Boston, MA 02125-3393, 617-287-6619, 6684; E-mail:
[email protected] Dr. Stephen M. Sagar: Departments of Oncology and Medicine, McMaster University, Hamilton, Ontario, Radiation Oncologist, Juravinski Regional Cancer Centre, Hamilton Health Sciences Centre. Associate Brain-Body Institute (St Josephs Health Care Centre and McMaster University); E-mail:
[email protected] Dr. Anil K. Verma: Sr. Asstt. Professor of Zoology, Govt. (P.G.) College for Women, Gandhi Nagar, Jammu-180 001(J&K State), India; E-mail:
[email protected] Prof. Ian Fraser Pryme: Dept. of Biomedicine, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway; E-mail:
[email protected] Dr. Robert Frangez: Institute of Physiology, Pharmacology and Toxicology, Veterinary Faculty, University of Ljubljana, Slovenia; E-mail:
[email protected] Dr. George Qian Li: Herbal Medicines Research and Education Centre Faculty of Pharmacy, The University of Sydney, NSW 2006, Australia, E-mail:
[email protected] Prof. Yuji Nagashima: Department of Food Sciences and Technology, Tokyo University of Marine Sciences and Technology, Konan 4-5-7, Minato, Tokyo 108-8477, Japan; E-mail:
[email protected] Prof. Pius Mpiana Tshimankinda: Departement de Chimie, Faculte des Sciences, Universite de Kinshasa, B.P. 190, Kinshasa XI, RD Congo; E-mail:
[email protected] Prof. Supayang Piyawan Voravuthikunchai: Natural Products Research Center and Department of Microbiology, Faculty of Science, Prince of Songkla University, Hatyai, Songkla, Thailand - 90112; E-mail:
[email protected]
SERIES ISBN: 1-933699-50-7
About the Series Nature abounds with a rich potential heritage ofbioactive natural products as that have been exploited for effective and beneficial use either as a therapeutic resource against a human disease or as a chemopreventive, antioxidant, nutraceutical or diet supplement. A natural product is a substance or a chemical compound produced by a living organism found in nature that usually has a pharmacological or biological activity. Some of these agents are derived from terrestrial plants, whereas, others are obtained from microorganisms, marine or freshwater organisms and even the higher animals. Past few decades have witnessed an unprecedented worldwide growth in the interest of bioactive natural products based medicines, both in developed and developing countries, and offer a resource-pool for lead identification in drug discovery owing to the reason that isolation of a novel molecule is easier than de novo synthesis, especially when the molecular structure of the compound is very complex. On account of such advantages, pharmaceutical companies are opting for bioactive natural products based drugs. More than 20 new drugs, launched world over between 2000 and 2005, originate from natural products. Scrutiny of medical indications by source of compounds has demonstrated that natural products and related drugs are used to treat 87% of all categorized human diseases (infectious and non-infectious). The development of new bioassay techniques, biotechnology methods, screening and high performance analytical methods have introduced new concepts and possibilities of rational drug design and drug discovery. Interest in bioactive natural products research is growing more strongly and can be attributed to several factors, including remarkable diversity of natural products, unmet therapeutic needs, the development of novel and sensitive techniques to detect, isolate, purify and structurally characterize these active constituents. This has opened opportunities for multidisciplinary research that joins the forces of natural product chemistry, molecular and cellular biology, synthetic and analytical chemistry, bio-chemistry, biotechnology, general biology, and pharmacology to exploit the vast diversity of bioactive natural products. Taking cognizance of these facts, we have initiated efforts in editing the present series "Comprehensive Bioactive Natural Products" and brought eight volumes (1 to 8) providing edited information from over 139 original research and review articles by eminent scientists and researchers
from India and abroad, representing 40 countries ofthe world on wide range oftopics in the area of natural products categorized under the themes viz., 1. Potential & Challenges 2. Efficacy, Safety & Clinical Evaluation I 3. Efficacy, Safety & Clinical Evaluation II 4. Antioxidants & Nutraceuticals 5. Immune-modulation & Vaccine Adjuvants 6. Extraction, Isolation & Characterization 7. Structural Modifications & Drug Development 8. Quality Control & Standardization
These volumes critically evaluate the present state-of-art, current status and future prospects in a well-illustrated manner. It is hoped that this series, which reflects the contributor's research results as well as world wide reviews will be widely read and used by all interested in these fields and shall open new vistas of research and academic pursuits for those engaged in the fields of bioactive natural products. I would like to take this opportunity to thank each of the authors who have contributed in the preparation of this book series.
Jammu, India V.K. Gupta Series Editor
\ \11 I ) Ii
•
",
AMITY INSTITUTE FOR HERBAL AND BIOTECH PRODUCTS DEVELOPMENT - An Institution of Ritnand Balved Education Foundation - Thiruvananthapuram
Prof. (Dr.) P. PUSHPANGADAN, M.Sc. M.Phil. Ph.D., FBRS FES. FNRS, FNSE, FNESA, FNAASc, FNASc., (UN Equator Initiative Laureate) Director General & Senior Vice President, RBEF (Former Director, NBRI, Lucknow)
08-06-2009
Foreword to the Series Medicine among the ancient civilizations was a blend of Religion and Science. Natural products played an important role throughout the world in the prevention and treatment of diseases. These natural products originated from varied sources such as plants, animals, micro-organisms, marine organisms, terrestrial vertebrates and invertebrates. In man's quest for discovering new and potent remedies for diseases, he keenly observed his immediate neighbourhood and experimented not only with organic substances but also with in-organic materials such as metals, minerals, ores etc. Over the years, the accumulated medical wisdom was compiled into excellent treatises and these have later developed into the various classical systems of medicine associated with the ancient civilizations and cultures of the world. Apart from the classical systems of medicines, which by its nature and practice had universal applications, there existed another stream of medicine in the countryside, among the less privileged class of people, among the forest dwellers and among the nomadic tribes which were location specific, confined to small communities, tribes and families. These medical systems were mostly perpetuated by oral traditions. Nevertheless, the value of the oral traditions cannot be overlooked as these localized knowledge systems are veritable treasures of invaluable information on the properties and uses of hundreds of plant, animal and mineral products. Ethno medical information is now increasingly being utilized in the search for novel bioactive molecules from natural products. Several well known plants such as Licorice (Glycyrrhiza glabra) myrrh (Commiphora sps) and poppy capsule latex (Papaver somniferum) were mentioned in the clay tablets dating back to 2600 B.C, obtained from Mesopotamia. These plants and their derivatives are still used in various forms as herbal drugs as well as pure isolates in different systems of medicine. For instance,
codeine, papaverine, morphine etc isolated from P. somniferum are still used in modem medicine. Similarly hemisuccinate carbenoxolone sodium, a semi synthetic derivative of glycyrrhetic acid isolated from licorice is used for the treatment of gastric and duodenal ulcers. Plants, especially those with ethno medical history have been the major source ofbio active molecules. According to a study by Fabricant and Farnsworth (2001), out of 122 plant derived drugs, 80% had their origin from plants with ethno medical use. Since secondary metabolites from plants are products ofbiosynthetic reactions occurring in living systems, bioactive molecules from natural sources are excellent drug candidates as they are perceived to show more biological friendliness than synthetic drugs. The ever increasing popularity and acceptance of natural products have evinced an extra ordinary interest and scope in the subject in recent years. Compendium of Bioactive Natural Products- a series of eight books edited by Dr. V.K Gupta (Volumes 1,2,3 and 5) and Dr. V.K Gupta, Dr. S.C. Taneja, Dr. B.D. Gupta and Dr. A.K.Verma (Volumes 4,6,7 and 8), published by MIS. Studium press LLC, U.S.A are an excellent compilation, giving comprehensive and reliable data on the current development in the field ofbioactive natural products. This series contain 139 original research and review articles contributed by a galaxy of eminent scientists and academicians from 40 countries around the world. The editors have taken care to invite eminent scholars in the relevant areas to contribute each chapter in the series. The series deals with wide ranging topics related to natural products such as extraction, isolation and characterization, structural modifications and drug development, quality control and standardization and, immune modulation and vaccine adjuvants, antioxidants and nutraceuticals, efficacy, safety and clinical evaluation (Parts-1 and 2) and potentials and challenges. The editors of this series Dr. V.K Gupta, Dr. S.C. Taneja, Dr. B.D. Gupta and Dr. Anil K Verma are known to me for more than three decades and they are all stalwarts in natural products and related areas. It is gratifying to know that this series is dedicated to the memory of Col. Sir Ram Nath Chopra, the Founder Director of Regional Research Laboratory, now known as Indian Institute of Integrative Medicine (IIIM), Jammu.
Col. Sir R.N. Chopra (1882-1973) was a pioneer of systematic studies of indigenous drugs, promoter of Indian Systems of Medicine and Patron of Pharmacy in India. Most of the medicinal plants, he studied were in use, in Ayurveda for thousands of years. The major fields of Cot. Chopra's research were general pharmacology, chemotherapy, indigenous drugs, drug addiction and drug assays. At the Calcutta school of Tropical Medicine, he developed a pattern of research in studying the action of medicinal plants which influenced an entire generation of pharmacologists. Pharmacists and physiologists in India. In the words of Dr. G.V. Satyawati, Former Director General ofIndian Council of Medical Research, the credit of kindling the interest of Indian Chemists and Pharmacologists in Medicinal Plants should rightfully go to
Sir Ram Nath Chopra who, has been claimed as the Father of Indian Pharmacology' . I had the privilege and good fortune of meeting Col, Sir. Ram Nath Chopra along with Prof. Dr. E.K. Janaky Aromal at his residence in 1969 immediately after my joining Regional Research Laboratory as a Scientific Assistant. My meeting with Col. Sir R.N. Chopra had left a lasting impression of a great scientist in my memory. It is a fitting tribute to Col. Sir. R.N. Chopra that the editors have decided to dedicate these eight volumes to his memory. I congratulate the editors for bringing out this series which is timely, relevant and important to all those who are engaged in the study of natural products and development of therapeutically useful bioactive molecules. I am sanguine that these volumes will receive wider acceptance among the scientific community, especially among those engaged in Natural Product research.
(P. Pushpangadan)
About the Editor Dr. Vijay Kumar Gupta, Ph.D., FLS, London (born 1953-) Deputy Director & Head, Animal House, Indian Institute ofIntegrative Medicine (CSIR), Jammu, India. He did his M.Sc. (1975) and Ph.D. (1979) in Zoology both from University of Jammu, Jammu-India. His research capabilities are substantiated by his excellent work on histopathology, ecology and reproductive biology of fishes, turtles, birds and mammals, which has already got recognition in India and abroad. Dr. Gupta has to his credit more than 75 scientific publications and review articles which have appeared in internationally recognized Indian and foreign journals. Founder fellow, life member and office bearer of many national societies, academies and associations. He has successfully completed a number of research/consultancy projects funded by various governments, private and multinational agencies. His current areas of interest are histopathology, toxicology, pre-clinical safety pharmacology and reproductive efficacy studies oflaboratory animals. He is also Editor-in-chief of the books 1) Utilisation and Management of Medicinal Plants 2) Medicinal Plants: Phytochemistry, Pharmacology and Therapeutics 3) Perspectives in Animal Ecology and Reproduction (Vols.1-6). The Editor-in-chief of the American Biographical Institute, USA, has appointed him as Consulting Editor of The Contemporary Who's Who. Dr. Gupta also appointed as Nominee for the Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA, Govt. oflndia). Recently the Linnaean Society ofLondon, U.K. has awarded fellowship to him in November 2009 in recognition of his contribution towards the cultivation of knowledge in Science of Natural History.
Preface The natural products have been of great help to mankind in resolving many diseases from earliest time. Primitive people in all ages have had some knowledge of natural medicines. The knowledge of the primitive man has been so modified with the advancement of civilization that know-a-days almost every type of medicine be that of Ayurvedic, Unani or even a number of allopathic medicines are obtained from natural sources. The medicinal value of drug plants is due to the presence of some chemical compounds in the plant tissue that produce a definite physiological action on human body. Early societies learned through trial and error that many plants contained substances with significant curative properties and over the years the natural products became used in more refined ways leading ultimately to the use of pure single component "active ingredient" as drugs. Nature has continuously provided mankind with a broad and structurally diverse array of pharmacologically active compounds that have proved to be indispensable for the cure of deadly diseases or as lead structures for novel pharmaceuticals. Their dominant role is evident in the approximately 60% of anticancer compounds and 75% of drugs for infectious diseases that are either natural products or natural product derivatives. At present 25% of the modern medicines are developed from plants that were first used traditionally, and many synthetic drugs have also been obtained from natural precursors. Almost, 70% modern medicines in India are derived from natural products. In the USA, the process of synthetic drug discovery and development takes an average of 12 years, and any new drug requires the investment of an average of US$ 230 million. It is seen that natural drugs take a comparatively much less time and expenses than synthetic drugs. Hence natural medicines would be cheaper, unless the market price are inflated by other considerations. Despite this success, during the past couple of decades, research into natural products has experienced a steady global decline. The introduction of high-throughput synthesis and combinatorial chemistry with their promise of a seemingly inexhaustible supply of compound libraries has greatly contributed to this declining interest in the screening of natural products by the pharmaceutical industry. Resistance of the parasites to existing drugs and their higher cost warrants the search of newer drug molecules. The origin of many effective drugs is found in the traditional medicine practices and in view of this several researchers have undertaken studies to clinically evaluate the medicinal plants for their preclinical safety and efficacy. The natural product market has been continually growing at an increased rate mainly as a nutraceuticals, dietary supplements, cosmoceuticals, chemopreventives and as herbal remedies. These therapies should emphasis on well-controlled and randomized clinical trials to prove
safety and efficacy and necessary guidelines should be followed in order to harmonies the use of bioactive natural products. At present, there is limited data on safety, efficacy and clinical trials to prescribe natural products in therapy. Realizing the need to document this knowledge gained through recent researchers, the present volume "Efficacy, Safety & Clinical Evaluation-I" of the book series entitled, "Comprehensive Bioactive Natural Products" presents 23 chapters discussing therapeutic potential of a large number of bioactive natural products of plant/animal origin investigated worldwide. Some of the interesting chapters included in this volume are: Perspectives in efficacy, safety and clinical evaluation of bioactive natural products; The modern application of traditional Chinese medicine: indigo naturalis as an alternative therapy in psoriasis vulgaris; Development of anti-angiogenesis therapies for treating cancer from Chinese medicinal herbs; Inflammation and mediators; Recommendation for reporting randomized controlled trials of herbal interventions: CONSORT for herbal medicine trials; Anxiolytic activity screening studies on extracts of a few medicinal plants; Larvicidal and antimicrobial activities of seeds ofAnnona cornifolia; Antidepressant activity of the extracts of three Hypericum species native to China; Cytolytic and toxic effects of ostreolysin, a protein from the oyster mushroom CPleurotus ostreatus); Antioxidant, antisecretory and gastroprotective activities from Leiothrix fiavescens; Evaluation of the immunity potency and security of inactivated oil-adjuvant vaccine against chlamydiosis in dairy cattle; Pharmacokinetics of active constituents of Rhodiola rosea SHR-5 extract; Metabolism and pharmacokinetic studies of coumarins; N aringin and its aglycone, research and managements; Biotransformation of drugs; Green tea: molecular targets in glucose uptake, inflammation, and insulin signaling pathways; Euphorbious plants as molluscicides and piscicides. The studies included are likely to lead further researches in this direction and it is hoped that this publication would attract world wide audience of researchers and the academicians of allied disciplines engaged in the search of new drug from natural resources.
Jammu, India
V.K.Gupta
ISBN: 1-933699-52-3
Table of Contents About the Series Foreword to the Series Preface Table of Contents of Volume 2 1.
Perspectives in Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
un
ix Xln
xu
1-29
YOGENDRANAYAK, VEERESHP. VEERAPUR, ANANTHA NAIK N AGAPPA AND M.K UNNIKRISHNAN (INDIA)
2.
The Modern Application of Traditional Chinese Medicine: Indigo N aturalis as an Alternative Therapy in Psoriasis vulgaris
31-43
YIN-Ku LIN AND JONG-HWEI Su PANG (TAIWAN)
3.
Development of Anti-Angiogenesis Therapies for Treating Cancer from Chinese Medicinal Herbs
45-65
STEPHEN M. SAGAR, RAIMOND K WONG AND DONALD R. YANCE JR. (CANADA, OREGON)
4.
An Anthology of the Studies on the Anti-Inflammatory, Antinociceptive and Febrifuge Effects of Medicinal Plants in Turkey
67-119
ESRA KUPEU AKKOL AND ERDEM YESILADA (TuRKEy)
5.
Comparative Study of the Antigenotoxic Effects of Some Selected Natural Plant Products Against Methylmethane Sulphonate Induced Genotoxic Damage in Cultured Mammalian Cells
121-131
YASIR HASAN SIDDIQUE AND MOHAMMAD AFZAL (INDIA)
6.
Inflammation and Mediators
A. KOUL, R. SHARMA, G.D. SINGH, A. KHAJuRIA AND V.K GUPTA (INDIA)
S. SINGH, P. KoUL,
133-178
7.
Recommendations for Reporting Randomized Controlled Trials of Herbal Interventions: CONSORT for Herbal Medicine Trials JOELJ. GAGNIER N.D., HEATHER BOON, PAULA ROCHON, DAVID MOHER, JOANNE BARNES AND CLAIRE BOMBARDIER FOR THE CONSORT GROUP (CANADA)
179-209
8.
Anxiolytic Activity Screening Studies on Extracts of a Few Medicinal Plants RICHA SHRI, MANJEET SINGH AND ANUPAM SHARMA (INDIA)
211-218
9.
Study of Eugeniajambolana and Momordica charantia in Respect to Safety and Antidiabetic Effect In vivo and In vitro YELE, SANTOSH AND VEERANJANEYULU, A. (INDIA)
219-230
10. Larvicidal and Antimicrobial Activities of Seeds of Annona cornifolia A. St. -Hil. (Annonaceae)
231-237
LUCIANA ALVES R.S. LIMA, MARIA AMELIA D. BOAVENTURA, JACQUELINE A. TAKAHASHI AND LUCIA P.S. PIMENTA (BRAZIL) 11. Antidepressant Activity of the Extracts of Three Hypericum species Native to China DEPO YANG, JIE BAr, ZHEN LI, GEORGE Q LI AND DONGMEI WANG (CHINA, AUSTRALIA)
239-249
12. Cytolytic and Toxic Effects of Ostreolysin, a Protein from the Oyster Mushroom (Pleurotus ostreatus)
251-264
KRIsTINA SEPCIC AND ROBERT FRANGE (REPUBLIC OF SLOVENIA) 13. Antioxidant, Antisecretory and Gastroprotective Activities from Leiothrix flavescens
265-279
THIAGO DE MELLO MORAEs, MARCELO APARECIDO SILVA, CLENILSON MARTINS RODRIGUES, LoURDES CAMPANER DOS SANTOS, MIRIAM SANNOMIYA, LUCIA REGINA MACHADO DA ROCHA, ALBA REGINA MONTEIRO SOUZA BRITO, TArS M. BAUAB, WAGNER VILEGAS AND CLELIAAKrKO HIRUMA-LIMA (BRAZIL) 14. Evaluation of the Immunity Potency and Security of Inactivated Oil-Adjuvant Vaccine against Chlamydiosis in Dairy Cattle Qru C., ZHOU J., CAO X. AND LIN G. (CHINA)
281-291
15. Annona crassiflora Wood Constituents: Antimalarial Efficacy, Larvicidal and Antimicrobial Activity
293-305
MARy ANE GONQALVES, VANESSA CARLA FURTADO MOSQUEIRA
AND
LUCIA PINHEIRO SANTOS PIMENTA (BRAZIL)
16. Pharmacokinetics of Active Constituents of Rhodiola rosea SHR-5 Extract A PANOSSIAN, A HOVHANNISYAN, H. ABRAHAMYAN, E. GABRIELYAN AND G. WIKMAN (ARMENIA, SWEDEN)
307-329
17. Metabolism and Pharmacokinetic Studies of Coumarins BEHERA D. AND AnDEPALLI V. (INDIA)
331-353
18. N aringin and its Aglycone, Research and Managements RICKYW.K WONG AND FANNYY.F. YOUNG (HONG KONG)
355-387
19. Biotransformation of Drugs M.G. TYAGI, S. PRABHAKARAN AND KR. DHANASEKHAR (INDIA)
389-398
20. Pharmacokinetics and Drug Interactions of Glycyrrhizin/Glycyrrhiza AK AGGARWAL AND D.K MAJuMDAR (INDIA)
399-416
21. Determination of Total Antioxidant Status and Rosmarinic Acid from Orthosiphon stamineus in Rat Plasma GABRIEL AKYIREM AKOWUAH AND ISMAIL ZHARI (MALAYSIA)
417-427
22. Green Tea: Molecular Targets in Glucose Uptake, Inflammation, and Insulin Signaling Pathways HEPING CAO, ANNE M. ROUSSEL AND RICHARD A ANDERSON (USA, FRANCE)
429-448
23. Euphorbious Plants as Molluscicides and Piscicides: A Review RAM P. YADAV AND AJAY SINGH (INDIA) Appendix
449-460
Table of Contents of Other Volumes of the SeriesVois. 1 & 3 to 8
461-474
Index
475-493
"This page is Intentionally Left Blank"
1 Perspectives in Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products YOGENDRA NAYAK1, VEERESH P. VEERAPUR2 , ANANTHA NAIK NAGAPPA3 AND M.K UNNIKRISHNAN 1
*
ABSTRACT Natural products (NPs) include crude drugs, extracts, botanicals and pure compounds obtained from nature. NPs offer a rich source of diverse biologically active compounds with high molecular diversity, varying structural complexity and unparalleled novelty. A significant portion of the currently prescribed drugs owe their origin either directly or indirectly to NPs. The bioactivity of NPs not only builds the foundation for ethnic medicine systems of the world but also provides an impetus to modern drug discovery. Isolation of active component / molecule, structure elucidation, biological evaluation, dereplication, semisynthesis, biosynthesis, as well as optimization of downstream processing have undergone revolutionary changes in recent times, thereby enhancing the rate of drug discovery. The advent of high throughput screening and the availability of combinatorial compound libraries in natural products have been attracting major innovators towards natural products. Translational research has brought the drug discovery team closer to the clinical setting and has simultaneously increased the collective involvement of clinicians and pharmaceutical scientists. Complexity of natural products is fitly matched by the complexity 1. Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal- 576 104, India. 2. Department of Pharmaceutical Chemistry, SET's College of Pharmacy, S.R. Nagar, Dharwad - 580 002, India. 3. Department of Pharmaceutical Management, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal- 576 104, India. * Corresponding author : E-mail:
[email protected]
2
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
of several pathological conditions, for instance diabetes, where multiple drug targets work in unison for producing a therapeutic outcome. Reverse pharmacology makes use of clinical evidences from traditional medicines, and then working backwards towards validation of claims in the laboratory. All the above themes are discussed in detail. Key words: Combinatorial compound library, drug discovery, high throughput screening, multiple drug targets, natural products, reverse pharmacology, translational research
IMPACT OF NATURAL PRODUCTS ON DRUG DISCOVERY: HISTORICAL PERSPECTIVES Natural products are rich sources of biologically active compounds with tremendous molecular diversity, possessing great promise in drug discovery and development (Butler, 2004; Newman et al., 2003a; Koehn et al., 2005; 2004b; Chin et al., 2006; Cragg et al., 2006; Blunt et al., 2007; Gordaliza, 2007; Butler et al., 2008). There is no dearth of illustrative examples to prove this paradigm. One of the oldest drugs, aspirin has had its origin from a plant (bark of willow), making it a historical land mark (Vane, 2000; Robert-II et al., 2001). Penicillin was first isolated from a microorganism Penicillum notatum (Wainwright, 1990). Morphine obtained from opium poppy is still an important drug in pain management. Bergmann's discovery and subsequent identification of spongothymidine and spongouridine in the early 1950s from the Caribbean sponge Tethya crypta led to the development of a nucleoside with biological activity (Bergmann, 1950). An explosive growth in synthetic chemistry further led to the discovery of a close analogue, cytosine arabinoside, a potent antileukemic agent (Beranek, 1986; Suckling, 1991). This compound was subsequently commercialized by Upjohn (now Pharmacia) as Ara-C. A closely related compound adenine arabinoside (Ara-A) was later synthesized and commercialized by Burroughs Wellcome (now Glaxo Smith Kline). Similarly, insulin was first isolated from the pancreatic extracts of animals such as cows, pigs and sheep, till recombinant DNA technology replaced the old technology. Eli Lilly (Indianapolis, USA) later demonstrated the commercial feasibility of recombinant DNA technology with the introduction of human insulin in 1982, ushering in a paradigm shift in drug discovery and development, leading to the start of the 'biotech era' (Pillai, 2001). The discovery of cyclosporine from microbial sources revolutionized immunosuppressive therapy for organ transplant patients (Kahan, 1999). Indeed, it has been estimated that at least 60% of the roughly 1184 new therapeutic agents introduced world-wide over the last twenty five years (1981-2006) have had their origins from natural products in one way or the other (Newman et al., 2007). The changing strategies and innovations in NP research have greatly influenced drug discovery programmes, in which
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
3
sample selection and collection, processing, isolation of active componenti molecule, structure elucidation, biological evaluation, dereplication, semisynthesis, biosynthesis, as well as optimization of downstream processing has become the norm. Such strategies have substantially enhanced the rate of discovery of novel NP with renewed interest in recent years (Gullo et al., 2005; Lawrence, 2005a, 2007b). With the advance of high throughput screening (HTS) and new instrumentation, the discovery of the new molecules with biological activity has hastened further. Newer guidelines and statutes implemented by various regulatory authorities have enhanced the time frame for the assessment of safety and efficacy of drugs in human populations. It can take up to fifteen years for the development of a molecule before it is commercialized into a marketed drug. Apart from the passage of time, a great amount of money is being spent in the process of developing a product into a clinically useful drug (Frank, 2003; Schmid et al., 2007). It is therefore important to examine the various events in drug discovery from natural sources and the associated regulatory rigors in assessment of quality, efficacy and safety of the drug. It is also essential to look into the translational aspects of NPs from preclinical to clinical studies (Dickson et al., 2004). The recent increase in the development-costs of new chemical entities (NCEs) has forced many innovators to search for active drug molecules from nature. The vast reservoir of diverse sources and the infinite variety in nature's molecular repertoire offers a fascinating arena for the drug discovery team.
NATURAL PRODUCTS FROM PLANT ORIGIN Medicines from plant sources have been used since ancient times for a variety of ailments. Herbal preparations have been used in the art of healing since time immemorial. Primitive man, through trial and error, gained knowledge of herbals and passed it on to their progeny. It appears that for thousands of years herbs were perhaps used for both their magical powers as well as medicinal values (Oumeish, 1998). Ancient civilization flourished in 3000 BC onward in Egypt, Middle East, India and China, with a parallel growth in the refinement of herbal medicine. The Egyptian Ebers Papyrus (1500 BC) is one of the earliest records providing information on herbs (Frey, 1986; EI-Gammal, 1994). In India, circa 1500 BC, Vedas included the Ayurveda system of medicine along with its vastly sophisticated informationbased on herbs, with about 350 herbals cited in this compilation (Patwardhan et al., 2004). Chinese traditional pharmacopoeia lists over 5700 traditional medicines which are mostly of herbal origin (Qiu, 2007). Chinese traditional medicine now enjoys credibility that is comparable to Western medicine in many parts of the world. Many universities in China teach and practice herbal medicine. Over the last 10 years Europe has witnessed a growth in the practice of Traditional Chinese Herbal Medicine.
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
4
The development of organic chemistry has shown that the ability of herbal medicine to treat the body depended upon its chemical constituents. Chemists first began extracting and isolating chemicals from plants in the 18 th century. Encouraging results accelerated their enterprise and NP chemists have given to the world some of its most useful drugs such as morphine, digoxin, d-tubocurarine, ephedrine, quinine, vincristine, paclitaxel etc. A detailed list is presented in Table 1. Table 1. Prescription drugs derived from natural sources
Use Amino glycoside antibiotic
Analgesic
Drug name and the source Streptomycin (Bacteria; Streptomyces griseus) Gentamicin (Micromonospora) Kanamycin (Streptomyces kanamyceticus) Neomycin (Streptomyces fradiae) Netilimicin sulfate (Micromonospora) Morphine (Fruit exudate; Papaver somniferum)
Non-opioid analgesic
Epibatidine (Alkaloid that originally is found in the skin of a neotropical poisonous frog, Epipedobates tricolor); 200 times stronger than morphine; derivatives are under clinical investigation
Ansamycin antibiotic;
Rifamycin (Amycolatopsis rifamycinica); Anti-TB
Anti-Alzheimer's
Galantamine hydrobromide (An alkaloid obtained from Galanthus nivalis); Selective acetylcholinesterase inhibitor
Antibiotic
Mupirocin (Mixture of several pseudomonic acids obtained from Pseudomonas fluorescens); active against methicillin-resistant Staphylococcus aureus (MRSA) Chloramphenicol (bacterium; Streptomyces venezuelae) Tetracycline (polyketide antibiotic from Streptomyces) Daptomycin (Streptomyces roseosporus); Cyclic lipopeptide antibacterial agent for skin infections Teicoplanin (Actinoplanes teichomyceticus); Glycopeptide antiobiotic active against MRSA Vancomycin (Amycolatopsis orientalis); Glycopeptide antibiotic active against penicillin resistant Staphylococcus aureus Lincomycin (Streptomyces lincolnensis); Lincosamide antibiotic Erythromycin (actinomycete, Saccharopolyspora erythraea); Macrolide antibiotic Aztreonam (Chromobacterium violaceum); Monobactam antibiotic Cephalosporin (fungus; Cephalosporium acremonium); Beta lactum antibiotic Penicillin (Fungus; Penicillum notatum); Beta lactum antibiotic Colistin (polymyxin E) (Bacillus polymyxa var. colistinus); Polymyxin antibiotic Bacitracin (Bacillus subtilis var Tracy); Polypeptide antibiotic Fusidic acid (fungus: Fusidium coccineum); Urinary antibiotic
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
5
Table 1. Contd. Use
Drug name and the source
Anticancer
Arglabin (from a species of wormwood Artemisia glabella endemic to the Karaganda region of Kazakstan) Bleomycin (Glycopeptide antibiotic from bacterium: Streptomyces vertic ill us) Etoposide (Herb: podophylum). Masoprocol (Nordihydroguaiaretic acid; NDGA; is a potent antioxidant compound found in the long-lived creosote bush: Larrea tridentata) Paclitaxel (plant; Taxus brevifolia) Solamargines (plant; Solanum marginatum) Vinblastin, vincristine (herb; Vinca rosea)
Anticancer antibiotic
Daunorubicin (Streptomyces peucetius) Pentostatin (Streptomyces antibioticus) Daunomycin (from bacterium: Streptomyces peucetius) Doxorubicin (Adriamycin; related to daunomycin from new strain of Streptomyces peucetius) Mithramycin (Plicamycin; Streptomyces plicatus) Mitomycin C (Streptomyces lavendulae) Sarkomycin (Streptomyces erythrochromogenes).
Anticoagulant
Heparin (sheep liver)
Antidiabetic
Insulin (pig and sheep) Voglibose (Aspergillus awamori)
Antifertility
Diosgenin (rhizome; Dioscorea deltoideae)
Antihypertensive
Reserpine (Rauwolfia serpentina)
Antimalaria
Quinine (Cinchona bark) Artemisinin (herb, Artemisia Annua)
Antiplatelet
Bivalirudin anew, genetically engineered form of hirudin, the substance in the saliva of the leech (Haementeria officinalis); used to reduce the risk of blood clotting in unstable angina who are undergoing a procedure to open blocked arteries in the heart.
Anti-spasmodic
Ephedrine (herb: Ephedra)
Cardiotonic
Digoxin (Fox glove; Digitalis species)
Hypolipidemic
Mevastatin (fungus; Penicillium citrinum)
Immunosuppressant for organ transplant
Sirolimus (Rapamycin; from bacterium Streptomyces hygroscopicus) Tacrolimus (bacterium Streptomyces tsukubaensis) Cyclosprin (fungus; Tolypocladium inflatum)
Local anaesthetic
Cocaine (plant; Erythroxylum coca)
Muscle relaxant
d-tubocurarine (plant; Chondrodendron tomentosum)
Mydriatic
Atropine (herb; Atropa belladonna)
6
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
In spite of the great advances in synthetic and combinatorial chemistry, medicinal plants still make an important contribution in pharmaceutical innovation. According to the World Health Organization (WHO), about 65-80% of the world's population living in developing countries, still depends essentially on plants for primary health care because of poverty and lack of access to modern medicine (Pal et al., 2003). Some Western countries such as Germany, France, Italy and the United States have developed appropriate guidelines for registration of herbal medicines. Discovery of the blockbuster anticancer drug paclitaxel (Taxol) from the yew tree (Taxus brevifolia) once again helped to revive the interest towards herbal medicine (McChesney et al., 2007). Nowadays, several major pharmaceutical companies demonstrate renewed interest in investigating plants for discovering new sources for new lead structures and also for the development of standardized phytotherapeutic agents with improved efficacy, safety and quality (Gordaliza, 2007; Potterat et al., 2008).
NATURAL PRODUCTS FROM MARINE SOURCES Marine NPs have yielded a considerable number of new drug candidates which are still in preclinical or early clinical development (Haefner, 2003; Marris, 2006; Saleem et al., 2007). However a few are already in the market, such as cytarabine. Drugs from marine sources have a wide range of therapeutic benefits such as anticancer, antiviral, antiinfective, antimicrobial, analgesic, anti-inflammatory, anti-neurodegenerative in addition to many other therapeutic classes (Donia et al., 2003; Rawat et al., 2006; Bao et al., 2007; Diers et al., 2008; Uemura, 2006). Some of the NPs isolated from marine invertebrates have been shown to be, or are expected to be of microbial origin (Tan, 2007). Marine microorganisms, whose immense genetic and biochemical diversity is only beginning to be appreciated, appear to serve as a rich reservoir of NCEs. The prokaryotic marine cyanobacteria are a potential source of structurally bioactive secondary metabolites. A number of cytotoxic compounds such as hectochlorin, lyngbyabellins, apratoxins, and aurilides have been identified as potential lead compounds for the development of anticancer agents (Jimeno et al., 2004; Braiia et al., 2006).
NATURAL PRODUCTS FROM MICROORGANISM The seemingly endless potential of microorganisms dawned upon mankind in 1928 with Alexander Fleming's sensational discovery of an antibacterial filtrate "penicillin" from a commonplace mould Penicillium notatum (Ligon, 2004). Re-isolation and clinical studies pioneered by Chain, Florey, and co-workers in the early 1940s, and the subsequent scale-up towards commercially viable manufacture of synthetic penicillins revolutionized the foundation of antimicrobial drug discovery (Bentley, 2005). The
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
7
phenomenal success of penicillin encouraged drug companies and research groups to pool their resources together for a large scale culture of a diverse array of microorganism from which several newer antibiotics emerged (Knight et al., 2003). Early years of this large scale hunt for antibiotics yielded a number of successful antibiotics, some of which are widely prescribed to this day. Prominent among these include pathbreaking molecules in the history of modern medicine, such as streptomycin, chloramphenicol, tetracycline, cephalosporin, erythromycin, and vancomycin. Each one of these compounds, either in its original form or in its synthetic derivative, can be found on the shelves of a pharmacy to this day. The impact of these drugs has even served to change the very attitude towards infective diseases from one of horror and despair to that of a routine ailment of trivial consequence. More lives have been saved in history by antibiotics derived from nature, than through any other class of prescribed drugs. In other words, NP derived drug molecules have contributed the most towards increasing the life expectancy of man. Antibiotics of natural origin also remain some of the most often prescribed drugs in the world. Some of the earliest compounds discovered in the early 1970s using rational approaches based on mechanism of action, include the betalactamase inhibitor clavulanic acid from Streptomyces clavuligerus (Brown et al., 1976) and the HMG-CoA reductase inhibitor mevastatin (earlier code-named as ML-236B) from Penicillium citrinum (Endo, 1981). A mixture of clavulanic acid and amoxicillin (the combination is called Augmentin) is being used today as a front line antibiotic in several critical infections, while mevastatin and lovastatin, have been the leads in the discovery of a series of drugs in the treatment of dyslipidemia. Today, these groups of statins are among the top revenue earners for some of the richest drug companies in the world (Manzoni et al., 2002). The arrival of statins has also widened the scope of therapeutic possibilities in compounds isolated from microbes. Many biotech industries are now utilizing microorganisms as the raw material for the production of not only antibiotics, but also various enzymes, antibodies, vaccines and many genetic engineered hormones.
EXPLOITING NEW APPROACHES FOR NATURAL PRODUCT BY PHARMACEUTICAL COMPANIES Just as the arrival of aspirin revolutionised the preparation of medicines from an individual enterprise of laboratory dispensing into a scale of industrial manufacture, the invention of penicillin and other antibiotics ushered in a new era of large-scale manufacture through biotechnology. Although 60% of the new drugs owe its origin to NPs, this enormous pool of resources is being steadily overlooked by several large pharmaceutical innovators for over a decade (Baker et al., 2007). This strategy seems
8
Comp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
inappropriate because several synthetic analogues of the NPs have often proved themselves to be more efficacious and less toxic. For instance, chloroquine is less toxic than quinine, and homatropine is shorter-acting and therefore produces less cycloplegia than atropine. Lignocaine, the more therapeutically useful product, was produced by modifying the cocaine molecule. Semisynthetic penicillins like methicillin expanded the range of cure. Similarly, second and third generation synthetic cephalosporins have extended the antibacterial spectrum of the prototype. The advent of high throughput screening (HTS, see below) and the availability of combinatorial compound libraries have been attributed as major reasons for this change in attitude (Boldi, 2004). NP-based drug discovery has been finding it difficult in adapting itself to modern techniques such as HTS and rapid hit-identification process. This disadvantage is being felt most acutely by large pharmaceutical companies with enormous funds at their disposal. The long time-gap between drug discovery and its marketing tends to challenge the wisdom in this tendency to move away from natural products. However, the scenario will become clear only in the long term. Interestingly, smaller biotech companies are emerging with renewed strategies to focus on exploiting new approaches for NP-based drug discovery. These approaches include the application of genomic tools, identification of novel sources of organisms, newer technologies (listed in the Table 2) and improved processes of sample preparation for screening. In addition, the recent focus on the synthesis of diversity-oriented combinatorial libraries based on NP-like compounds is an attempt to enhance the productivity of synthetic chemical libraries (Marcaurelle et al., 2008). One of the breakthroughs in drug discovery was the use of mechanism-based screening for bioassay-guided fractionation (Phillipson et al., 2002). Efforts continue to improve screening formats, reagent production, robotics, and data management. Mechanism-based screening Table 2. Technologiestl'echniques applied in drug discovery from natural products Name of the technology! technique Chromatography: High performance liquid chromatography (HPLC) High performance thin layer chromatography (HPTLC) Gas chromatography (GC) Liquid chromatography (LC)
Applications Isolation of components from the natural sources. In vitro and in vivo drug metabolism and pharmacokinetic (DMPK) assays, HTS new molecule identification, Biomarker identification (Korfmacher, 2005; Castro-Perez, 2007)
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
9
Table 2. Contd. Name of the technology! technique
Applications
Supercritical fluid chromatography (SFC)
For the isolation of chiral compounds from natural sources (Zhang et al., 2005)
Mass Spectroscopy (MS): GC-MS, HPLC-MS, HPLCMS-MS, LC-MS, Fluorescence activated cell sorting (FACS)-MS, Liquid chromatography photodiode array mass spectrometry (LC-PDA-MS)
Molecular identification, and the structural elucidation of natural products usually combined with GC or HPLC or LC (Ma et al., 2006; Koehn, 2008)
FTICR-MS: Fourier transform-ion cyclotron resonance (FT-ICR) mass spectrometry
Biomarker discovery and analysis.Metabonomics, proteomics, natural product analysis and noncovalent complexes (including thermodynamics) (Feng et al., 2007; Zhang et al., 2005)
Nuclear magnetic resonance spectroscopy
Study on ligand binding to the protein, helps in the structure based drug design; Structural activity relationship (Robert, 2000; Coles et al., 2003; Pauli et al., 2005) Molecular isolation and identification usually combined with the HTS analysis (Koehn, 2008) For DMPK studies, HTS analysis of natural products (Corcoran et al., 2003) Studies of psychoactive agents in brain, Drug accumulation and metabolism in cancer, Quantitation and metabolism in the brain, liver and other organs (Paulus et al., 2007) To assess the biological function for unannotated proteins expressed by different genes with the protein structure solved (Reid et al., 2008; Powers et al., 2008)
(NMR)
Saturation Transfer Difference (STD)-NMR LC-NMR-MS, On-line solid-phase extraction (LC-SPE-NMR) Fluorine magnetic resonance (19F MR) Functional NMR (fNMR), Functional annotation screening technology by NMR (FAST-NMR) Positron emission tomography
Disease specific marker identification in nurological diseases (Wang et al., 2005; Beckmann et al., 2007; Hutchins et al., 2008)
High-throughput X-ray crystallography Wide-angle X-ray scattering
For protein structure studies, screening functional ligands, and ligand binding (Senior, 2005)
High-content highthroughput fluorescent microscopy
For the study of protein localization, dynamics and interactions with the natural products (Auer et al., 1998; Zemanova et al., 2003; Starkuviene et al., 2007)
has become the mainstay of HTS (Zheng et al., 2008). By utilizing new approaches, these companies are increasing the chemical diversity of their collections.
10
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
SCREENING OF NATURAL PRODUCTS Traditionally, crude extracts have been used routinely for screening. Aqueous or alcoholic extracts are most frequently used for the initial round of biological and phytochemical screening. Recently developed bioactivity-guided fractionation processes can now separate crude fractions into mixtures containing only a few compounds. Column chromatography, highperformance liquid chromatography (HPLC), countercurrent chromatography (CCC), or centrifugal partition chromatography (CPC) are some of the techniques used for fractionation (Maugh, 1983). Fractionation will allow active compounds present in amounts too small to detect, whose activity is masked in a crude extract. However, by screening at higher concentrations, interfering compounds will also be more frequently identified. The paradox is that the total number of fractionated extracts screened is inversely proportional to the biodiversity. A decision must be made whether the loss of biological diversity is adequately compensated by fractionation. The answer to this question will help determine the optimal number of fractions that should be generated for each extract. The advent of sophisticated separation by analytical instruments attached to the HTS enables scientists to assemble large libraries of pure NPs, which can then be screened in a manner analogous to pure compound libraries (Liu et al., 2004). A recent review by McChesney et al. (2007) discusses latest methods and techniques involved in the natural product extraction, isolation and purification. The identification of the compounds that are responsible for the activity of an extract is called dereplication process (Konishi et al., 2007). This process prioritizes extracts for further study and can save considerable research time. The most generic procedure used today involves separation of an extract using reversed-phase HPLC and splitting of the eluent post column into a mass spectrometer (usually MSIMS to match fragmentation ions) and a fraction collector using micro-titer plates to test tubes, depending on the scale (Strege, 1998). The fractions are screened and the retention time, UV spectra, MS data, and activity of fractions are analyzed for common interfering compounds and known inhibitors using commercial and in-house databases (Schuffenhauer et al., 2005). Other methods for dereplication that have been used include separation by solid-phase extraction cartridges and CCC. Alternative methods for analysis include affinity capillary electrophoresis, LC-NMR, and online biochemical assay (Bobzin et al., 2000; Beger, 2006). An increase in the speed of bioassay-guided fractionation has been facilitated by a marked improvement in HPLC automation, MS, and column technology, as well as rapid turnaround of screening results provided by HTS (Mishra et al., 2008). The advent of new probe technology and higher magnetic fields has led to a significant shortening in acquisition time for
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
11
NMR data, and the structure elucidation of NPs can be achieved routinely on amounts less than 1 mg (Liu et al., 2004; Xing et al., 2007). Automated structure-solving algorithms have made the job simpler but no technique can replace the structure elucidation skills of an experienced chemist (Ross et al., 2001). However, these programs add value to the tools that search for alternative structures that fit the same NMR data (Beger, 2006). IDGH THROUGH PUT SCREENING OF NATURAL PRODUCTS The discovery of truly novel NPs is a laborious and time consuming task when pursued through conventional programs such as extraction, fractionation, isolation and screening for biological activity. This has necessitated the adoption of high-throughput screening (HTS) by major players in the drug discovery team. HTS is defined as the automatic testing of potential drug candidates at a rate in excess of 10,000 compounds per week (Liu et al., 2004; Pereira et al., 2007; Koehn, 2008; Jenwitheesuk et al., 2008). HTS has gained widespread popularity over the last few years. The aim of high throughput drug discovery is to test large compound collections for potentially active compounds ('hits') in order to allow further development of compounds for pre-clinical testing ('leads'). In this approach, the molecular diversity and range of biological properties displayed by natural products is a challenge to combinatorial strategies for NPs synthesis and derivatisation (Posner, 2005). Many large companies study a hundred targets or more each year, and in order to validate these targets, lead compounds must be found. HTS is the process of assaying a large number of potential effectors of biological activity (lead) against the targets (a biological event) by miniaturized in vitro assays. Thus HTS technology has emerged over the last few years as an important tool for drug discovery and lead optimization in NP based drug discovery (Posner, 2005). HTS uses standard assay types known to most biological and biochemical scientists (e.g. ELISA, proliferation/cytotoxicity assays, reporter assays, and binding assays) (Inglese et al., 2007). The biochemical assay will be selected depending on the mechanism of action or target specificity of the compound or the mixture of compounds (commonly the fractionated extract). The primary assay is either cell free biochemical assays or cell based assays. Cell based assays allow the HTS to address a specific target (Castel et al., 2006). In order to process thousands of assays per day, HTS uses a combination of multiple well microplates and robotic processing. For a number of years, HTS assays have been run in the standard 96-well microplate (working volume of up to 250 ~L). The current goal of most companies is to move beyond this format to higher-density, lower-volume formats (e.g. 384 and 1536 well microplates). There are two primary advantages ofthese formats: increased throughput and lower volume, which
12
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
translates into lower cost. At screening rates of 500,000 compounds/week, even a cost of $11 well becomes too big a burden on the company's weekly budget (Mishra et al., 2008). The reduction of cost, rather than increase in throughput, is the primary driving force within most HTS groups to move towards higher-density, lower-volume microplates. Fluorescence polarization (FP) detector technology is widely acceptable in the plate reader system (Burke et al., 2003; Rogers, 1997). For example screening assay for detection of apoptosis through activated caspase is assessed as liberation of free rhodamine following incubation of treated cells with rhodamine-labeled (quenched) substrate solution. Compounds showing a five fold or greater increase in fluorescence are considered as hits, being inducers of caspase activity. However, intrinsic fluorescence of natural products can interfere with such detection. Use of special reagents such as Europium-labeled reagents and time resolved fluorescence detection, avoids this problem (McMahon et al., 2000).
COMPOUND DEVELOPMENT: TOTAL SYNTHESIS, SEMISYNTHESIS OR COMBINATORIAL SYNTHESIS Perhaps the biggest obstacle to NP chemistry is the supply oflarge amounts of NPs. For instance, a steady supply of paclitaxel from nature would require cutting down a large number of whole yew trees followed by tedious extraction of the active molecule (McChesney et al., 2007). A sustainable source of the NP should be identified whenever total synthesis is not possible. However, a microbial source does not face such a problem on account of the brief life cycle of the organism and efficient culturing methods sustainable in lab conditions. This is precisely the reason why most companies prefer screening of microbial products. Rapid advances in microbial combinatorial biosynthesis have hastened the process in recent times (Dougherty et al., 2006; Adrio et al., 2006). There has also been progress with organisms usually considered to be problematic. For example, the steady supply of compounds from plants can be enhanced with minimal environmental damage through tissue culture and genomic methods (Wilkinson et al., 2005; Chaturvedi et al., 2007; GOmez-Galera et al., 2007). The study is now being extended to cover aquaculture of marine invertebrates and the identification, cloning, and expression of symbiotic microbial genes (Munro et al., 1999). The molecular complexity of NP derived NCEs can also impede the lead optimization process. A very important trend in recent years has been a significant effort devoted to the semisynthesis and synthesis of complex NPs (Ortholand et al., 2004; Shang et al., 2005; Paul et al., 2007; Bulger et al., 2008) (Listed Table 3). The success of this strategy is evident from history. Literature
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
13
shows that a total of 322 NP-derived drugs have been launched from 1981 to 2006. More than 70% of these drugs (232 compounds) are manufactured by semisynthetic processes. Further, about 15% (47NCEs) have been produced by total synthesis (Newman et al., 2007; Butler, 2008). Paclitaxel, an anticancer drug manufactured by total synthesis, artemether, a semisynthetic drug from artemisinin, have emerged as synthetic breakthrough while vinblastine and vincristine (approved for cancer treatment in 1963 and 1965) templates are extremely difficult to synthesize economically. Table 3. Synthetic/semisynthetic analogues of natural products Name of drug
SourcelIndication
Acarbose
Hypoglycemic drug derivative of natural product from Aspergillus awamori
Alitretinoin
Anticancer; a first generation retinoid
Amrubicin
Derivative of doxorubicin from Streptomyces peucetius var caesius. Used in breast, lung, and gastric cancer
Anidulafungin
Antifungal agent; semi-synthetic lipopeptide from a fermentation product of Aspergillus nidulans.
Apomorphine hydrochloride
Synthetic analogue of morphine in Parkinson's disease
Arteether, artemether, artenusate
Semisynthetic derivative of artemesinin (from the plant Artemisia annual to treat malaria
Azithromycin
Macrolide antibiotic; Azane-substituent of erythromycin obtained from actinomycete Saccharopolyspora erythraea
Captopril, enalapril and cilazapril
Antihypertensive, developed from teprotide, isolated from Brazilian viper venom (Bothrops jararaca)
Carbepenams: Biapenem, Imipenam, Doripenem, Meropenam, Ertapenem Carumonam
Derived from a compound called thienamycin, produced by Streptomyces cattleya
Caspofungin acetate
Monobactam antibiotic, very resistant to ~-lactamase; derivative of aztreonam obtained from Chromobacterium violaceum Semisynthetic lipopeptide derived from pneumocandin BO, a fermentation product of Glarea lozoyensis; antifungal
Caspofungin acetate
Semisynthetic derivative of pneumocandin produced by fungi: Zalerion arboricola
Cefditoren pivoxil
An oral prodrug of cefditoren, a derivative of cephalosporin isolated from Cephalosporium species
Cladribine
Anticancer; related to nucleoside adenoside
Clari thromycin, Telithromycin, Dirithromycin
Macrolide antibiotic; Semi-synthetic erythromycin
14
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Table 3. Contd.
Name of drug Clindamycin Cytarabine ocfosfate
SourcelIndication Semisynthetic derivative of lincomycin, produced by the actinobacterium Streptomyces lincolnensis. Derivative of spongothymidine obtained from Caribbean sponge
Dicoumarol
Anticoagulant, derivative of plant coumarine isolated from fungal infected hay
Docetaxel
Anticancer; paclitaxel derivative
Ellipticine, Celiptium Epirubicin
Semisynthetic derivative from plant Ochrosia elliptica 4' -epimer of doxorubicin, an anticancer drug
Epirubicin HCI
Anticancer antibiotic related to doxorubicin
Etoposide phosphate
Anticancer; semisynthetic derivative from podophyllotoxin, found in the American Mayapple (Podophylum peltatuml
Everolimus
Orally active 40-0-(2-hydroxyethyl) derivative of rapamycin, originally produced from Streptomyces hygroscopicus.
Exemestane, Formestane
Anticancer; aromatase inhibitor structurally related to the natural substrate androstenedione
Exenatide
Antidiabetic: a synthetic analog of exenadin-4, which was originally isolated as a 39 amino acid peptide from the saliva of the Gila monster (Heloderma suspectuml
Fosfomycin trometamol
Antibacterial; analogue of fosfomycin obtained from Streptomyces fradiae.
Idarubicin hydrochloride (4demethoxydaunorubicinl
Anticancer antibiotic; derivative of daunourubicin
Irinotecan, topotecan
Camptothecin derivatives for cancer
Isepamicin
Aminoglycoside antibiotic; derivative of gentamicin B from Micromonospora, a genus of Gram-positive bacteria widely available in water and soil
Ivermectin
Anti-parasitic; derivative of avermectin obtained from microbe Streptomyces avermitilis
Micafungin sodium
Antifungal agent of the echinocandin type obtained from the culture broth of the fungus Coleophoma empetri ,and inhibits ~-(1, 3l-D-glucan synthase of fungi.
Micronomicin sulfate
Aminoglycoside antibiotic; 6'-N-methyl derivative of gentamicin C Antidiabetic; Acarbose related drug
Miglitol Miglustat
For type I Gaucher disease. An analogue of nojirimycin isolated from the broth filtrate of Streptomyces lavendulae
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
15
Table 3. Contd. Name of drug
SourcelIndication
Miokamycin
Derivative of macrolide antibiotic miodecamycin Al ; 9,3'diacetylmiodecamycin; has wide antimicrobial spectrum
Mitobronitol (1, 6-dibromo1, 6-dideoxy-d-mannitoll
Alkylating anticancer; derivative of mannitol
Mycophenolate sodium
Mycophenolic acid, originally purified from Penicillium brevicompactum, used for prophylaxis in renal transplant
Nandrolone phenylpropionate
Anticancer; Analogue of naturally occurring nandrolone in human
Nitisinone
Derivative of leptospermone, an important new class of herbicides from the bottlebrush plant (Callistemon citrinus)
Orlistatin
Antiobesity; Semisynthetic product from lipstatin (a natural product)
Peplomycin
Semisynthetic analogue of bleomycin from bacterium Streptomyces verticillus
Pimecrolimus
A novel analog of ascomycin, isolated as a fermentation product of Streptomyces hygroscopicus var ascomyceticus, for allergic contact dermatitis and atopic dermatitis
Pirarubicin
Anticancer antibiotic similar to daunorubicin
Rifampicin
Semisynthetic compound derived from rifamycin which is obtained from Amycolatopsis rifamycinica
Rosuvastatin calcium
Hypolipidemic drug; derivative ofmevastatin isolated from Penicillium citrznum and P. brevicompactum
Telithromycin
Semi-synthetic derivative of the 14-membered macrolide, erythromycin-A, isolated from Saccharopolyspora erythraea
Testolactone
Anticancer; derivative of progesterone
Tigecycline
9-tert-butyl-glycylamido derivative of minocycline, which is a semi-synthetic product of chlortetracycline isolated from Streptomyces aureofaciens.
Tiotropium bromide
Synthetic derivative of atropine from Atropa belladonna (Solanaceae) to treat COPD
Triproamylin acetate
Antidiabetic; amylin analog
Valrubicin
Anticancer antibiotic similar to doxorubicin
Vapreotide acetate
Anticancer; Somatostatin analogue
Vindesine and vinorelbine
Anticancer; semisynthetic derivatives of vinblastine from herb Vinca rosea
Ziconotide
Non opioid analgesic; synthetic form of one of the orconotoxins, obtained from cone snail (Conus magus) venom
16
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Synthetic effort has been particularly critical for NP-derived anticancer drugs where total synthesis has been crucial in lead optimization and for obtaining enough material for clinical trials. Synthetic chemists are actively engaged in such ventures where there are 15 derivatives of paclitaxel, 15 of camptothecin, 4 of combretastatin, 4 of vinca alkaloids and 2 of podophyllotoxin in the clinical trial process (Saklani et al., 2008). With the advent of combinatorial chemistry, a large number of compounds can be produced from a single lead discovered from the NPs (Lee et al., 2001; Nielsen, 2002; Boldi, 2004; Ganesan, 2004; Tan, 2004). Combinatorial libraries based on natural product scaffolds, analogs and derivatives are being currently pursued. For example, combinatorial chemistry techniques applied to scaffolds oftubulin inhibitors, have yielded second-generation paclitaxel analogs as anticancer drug candidates (Ladame et al., 2008). Further, compounds active against vancomycin-resistant bacteria have emerged from combinatorial libraries of vancomycin- dimers (Griffin et al., 2003). The combinatorial libraries of anticancer molecules designed to target receptor tyrosine kinase, rest on the evidences obtained from a natural product genistein (Kissau, 2003). Thousands of camptothecin derivatives have been synthesized with the combinatorial approach to target topoisomerase I. Topotecan and irinotecan are derivatives obtained in this manner (Li et al., 2006). In spite of all the sophistication, combinatorial chemistry has failed to deliver the high numbers of drug leads as was predicted. This is mainly because of the complexity and diversity of the NP scaffold. Limited success has paved way for "diversity oriented combinatorial synthesis" which proposes to produce molecules with greater structural complexity including enhanced stereo-chemical variations (Rouhi, 2003; Liao, 2003; Tan, 2005; Marcaurelle et al., 2008), but these approaches may have limited prospects in future compared to the natural product based combinatorial library (Koppitz et al., 2006).
TRANSLATIONAL RESEARCH IN ASSESSING SAFETY AND EFFICACY OF BIOACTIVE NATURAL PRODUCTS: FROM BENCH TO BED SIDE In drug discovery and development programs, a vast majority of tested compounds do not progress from laboratories to patients' bedsides. Historical evidence indicates that only 14% of the tested products entering phase I trials eventually obtain clearance from the FDA and enter the market (DiMasi et al., 2003). In recent times, chances of success have reduced to just 8% (Graul, 2007). This high attrition rate is being attributed to the knowledge gaps between in vitro data, preclinical data and the final clinical outcome (Eisenstein et al., 2008). The basic knowledge about the investigational molecule or the natural product is not being translated
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
17
appropriately to the clinical setting. The failures are not being analysed effectively together in a concerted manner by the drug discovery team and the clinical team. A two-way dialogue between scientists operating in preclinical drug discovery and clinical developers who understand the pathology and its alleviation, are required if novel leads and biologicals are to become useful drugs. Translational research, which focuses on such themes, has emerged as a new discipline in this context. The recent surge in translational research is expected to reduce the gap between the initial stages of drug discovery and subsequent clinical development (O'Connell et al., 2006). The American Physiological Society (APS) has defined translational research as 'the transfer of knowledge gained from basic research to new and improved methods of preventing, diagnosing or treating disease, as well as the transfer of clinical insights into hypotheses that can be tested and validated in the basic research laboratory (O'Connell et al., 2006). It is a kind of iterative learning experience from experimentation in basic and clinical fields. Thus in NP drug discovery, translational research interlinks preclinical and clinical research, where experiments progress steadily from lead generation towards proof of concept (early-stage clinical studies) (Sultana et al., 2007). Further, translational research assumes a broad perspective, embracing diverse areas such as clinical demands, public health policies and economic compulsions (Trusheim et al., 2007). The preclinical studies carried out in animals can predict efficacy (confidence in rationale) and safety (confidence in safety) of compounds or the natural product extracts in early development. The animal models and biomarkers (see below) can only give a preliminary idea of clinical correlation. Further, this may be poorly correlated to clinical outcomes in man. The pharmacokinetics and pharmacodynamics (PK-PD) of the compound in humans, which becomes clear only after the first dose in healthy volunteers, will help in determining the dose and route of administration. Research in normal human volunteers and patients enables a clearer understanding of the biology and pathophysiology, and can therefore trigger innovations in in vitro and animal experiments that better mimic the clinical condition. This strategy is therefore likely to enhance confidence in the appropriateness of a given target and help in dose selection for subsequent trials (Sartor, 2003). Biomarkers are quantitative measures of biological effects that provide informative links between mechanism of action and clinical effectiveness (Wehling, 2006). Assessment of biomarkers can reduce the uncertainty around key risks in the drug development cycle (pharmacology, PK-PD, toxicity, safety etc.). Further, biomarkers reflect biological effects induced by disease! test-compound and are the main tools to predict and describe its efficacy and safety. Specific markers like serum enzymes (ALT,
18
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
AST, Catalase, Alkaline phosphatase etc) and more general markers of disease-severity like C-reactive protein in inflammatory diseases are well known biomarkers. Histological parameters, imaging of tumors and the morphology of atherosclerosis, liver fat (safety biomarker), positron emission tomography (brain disorders) scans etc. are some disease-specific biomarkers (Colburn, 2003). In preclinical studies, death could be a biomarker in models of myocardial infarction, cancer, acute toxicity etc., being correlatable measures of efficacy. These biomarkers are used to achieve predictive power and this can vary from almost useless (e.g. serial brain slices etc., which can never be a human biomarker) to surrogate (e.g. LDL cholesterol, one of the few surrogates accepted by FDA), with most cases falling somewhere in between. Translational research initially was said to be bidirectional but it has got multidirectional approaches (Sonntag, 2005). It has an important role in identifYing new indications for established therapies - a process termed by some authors as 'indications discovery'. New drug target hypotheses can be generated through preclinical experiments on novel target organs. It can also result from clinical observations such as side effects or additional pharmacological effects beyond those expected from efficacy in the main indication (O'Connell et al., 2006). Sildenafil, a selective Phosphodiesterase5 inhibitor, is an example of a drug initially assessed for angina pectoris but was subsequently licensed for a completely different pathology; male erectile dysfunction and pulmonary artery hypertension (Ghofrani et al., 2006).
NATURAL PRODUCTS FOR TYPE 2 DIABETES: A CASE STUDY Diabetes mellitus is a group of metabolic diseases characterized by hyperglycemia resulting from defects in insulin secretion, insulin action, or both. The prevalence of diabetes for all age-groups worldwide was estimated to be 2.8% in 2000 and 4.4% in 2030. The total number of people with diabetes is projected to rise from 171 million in 2000 to 366 million in 2030 (Wild et al., 2004). Type 2 diabetes (T2D) is prevalent in 90% of the patients with diabetes. By 2025, more than three-quarters of all persons with diabetes will reside in developing countries. India and China are leading this surge in diabetes, and sub-Saharan Mrica is currently at a lower prevalence rate (Dagogo-Jack, 2006). Poorly managed diabetes leads to several complications (e.g. end-stage renal failure, blindness, amputation and heart disease) that many developing countries are ill equipped to tackle (Home, 2003). Diabetes is a heterogeneous metabolic disorder characterized by a decline in insulin action (insulin resistance), followed by pancreatic beta cell dysfunction (Chiasson et al., 2004). Insulin resistance will be associated with common conditions such as metabolic syndrome, dislipidaemia, obesity,
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
19
endocrine disorders and hypertension. Abnormal obesity, increased lipid profiles, atherosclerosis and hypertension are symptoms of the metabolic syndrome (Wild et al., 2007). Prescription drugs available today reduce blood glucose level either by increasing the tissue uptake or by increasing the insulin secretion. There is no successful drug to treat the metabolic complexes associated with diabetes (Matfin, 2008). A wide range of natural products are prescribed to the diabetic patient as nutraceuticals or dietary supplements for the intervention of disease, treatment and/or prevention (for more details http://www.naturaldatabase.com). Compounds of chromium, selenium, vanadium, herbal preparations of garlic, ginseng, a-lipoic acid and various mixed-herbal products are among the dietary supplements that are currently being used by many diabetics (Table 4). A recent review by Hays et al. (2008) discusses the rationale behind prescribing these natural products. Despite compelling evidence supporting an important role for dietary chromium in the restoration of normal glucose tolerance among chromium- deficient individuals, the ability of chromium to improve insulin action remains controversial (Balk et al., 2007). While a number of animal models have generally demonstrated anti-diabetic properties for ginseng, relatively few clinical studies in humans have been reported. Taken together, research data suggest that ginseng is likely to show a beneficial effect on certain metabolic parameters in T2D (Xie et al., 2005). However, it is difficult to make standardized clinical recommendations on account of the large reported variability in metabolic responses to different ginseng species, different batches of the same species, and different parts of the ginseng plant. On the whole, standardization of natural products remains a major bottleneck in the search for clinical evidence.
Table 4. Commonly used conventional and natural products for diabetes Prescription drugs
Natural products
Hypoglycemic agents Chlorpropamid Glimepiride Glipizide Glyburide Nateglinide Repaglinide Tolazamide Tolbutamide
Banaba (Lagerstroemia speciosal Bitter melon (Momordica charantial Fenugreek (TrigoneUa foenum-graecuml Gymnema (Gymnema sylvestre)
20
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Table 4. Contd. Prescription drugs
Natural Products
Insulin sensitizers Metformin Pioglitazone Rosigli tazone
American ginseng (Panax quinquefolius) Banaba (Lagerstroemia speciosa) Cassia cinnamon (Cinnamomum aromaticum) Chromium Ginseng (Panax ginseng) Prickly pear cactus (Opuntia ficus-indica) Soya (Glycine max) Vanadium
Carbohydrate absorption inhibitors Acarbose Miglitol
Miscellaneous Exenatide Pramlintide Sitaglitpin
Blond psyllium (Plantago ovata) Fenugreek (Trigonella foenum-graecum) Glucomannan (Amorphophallus konjac) Guar gum (Cyamopsis tetragonoloba) Oat bran (Avena sativa) Prickly pear cactus (Opuntia ficus-indica) Soy (Glycine max) a-lipoic acid Selenium Stevia (Stevia rebaudiana)
Numerous cell culture and in vivo animal studies have demonstrated that treatment with the water soluble antioxidant a-lipoic acid is associated with improved skeletal muscle glucose transport activity, reduced wholebody insulin resistance, and reduced oxidative stress. Although relatively few clinical trials have been reported, the combination of compelling in vitro and in vivo animal data in the presence of limited clinical data indicate the need for further research in human subjects while only supporting a general recommendation for a-lipoic acid supplementation (Bilska et al., 2005). Epidemiological studies on various populations living on a diet consisting of a high proportion of fatty fish (a rich source of omega-3 fatty acids) demonstrate a lower incidence of T2D compared to matched population groups. Positive changes in lipid profile and blood pressure demonstrated by epidemiological and other studies suggest that omega-3 fatty acid supplementation may reduce cardiovascular disease risk in T2D patients, even if no direct improvement in insulin or glucose homeostasis has been conclusively demonstrated (Lombardo et al., 2006). Many Indian medicinal plants have shown marked efficacy in the treatment of diabetes (Saxena et al., 2004; Modak et al., 2007). A recent review by MukheIjee et al. (2006) has discussed the Indian medicinal plants
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
21
with antidiabetic activity. However, detailed clinical investigations on the simultaneous use of prescription drugs along with natural products and dietary supplements are required for ensuring clinical safety and efficacy. A thorough preclinical and clinical investigation is necessary for establishing evidence. Diabetes presents a singularly complex condition where multiple targets operate in conjunction with the very complex physiology of nutrition and energy homeostasis. The number of druggable targets in diabetes is steadily increasing, with more and more successful drug classes entering the market. For instance, the DPP-IV (Dipeptidyl peptidase IV) inhibitor sitagliptin has recently been launched. While synthetic compounds address these targets individually, natural products probably tackle them in a collective and perhaps more holistic fashion. Restoring the equilibrium in the disturbed scenario of metabolic syndrome is probably linked to the ability to manage multiple targets taken together. While the effect on each individual target is sub-clinical, the collective pharmacodynamic picture is likely to playa role in restoring the complex metabolic equilibrium. For instance, we have demonstrated in our labs that extracts of Dodonaea I ~ t ~
f
SpeCific gene expression in
adlpocytes
.t- FA uptake '" Lipolysis
~
c==z:> ~ FFA
~oo~ t
II
- - _.......-
Insulin
'---V
sensitizer
.
factors
~ I
Expressionl action
.
"
~+
of insulin resistance factors
e.g. Reslstin! TNF
e ROS viz., ·OH, O2• •, Lpx Glucose Uptake
Fig 1. Schematic representation of the possible mechanisms by which a typical NP affords antidiabetic activity (PPAR y. Peroxisome Proliferator-Activated Receptor-Gamma; FA: Fatty Acids; FFA: Free Fatty Acids; TNF: Tumour Necrosis Factor; PTP-1B: Protein Tyrosine Phosphatase 1B; GLP-l: Glucagonlike peptide-1B; GIP: Gastric inhibitory polypeptide); DPP IV: Dipeptidyl peptidase IV; ROS: Reactive Oxygen Species; ·OH: Hydroxyl Radical; O 2 • -: Superoxide Radical; Lpx: Lipoxide Radicals)
22
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
viscosa and Ficus racemosa stem bark show antidiabetic activity through multimodal mechanisms in animal models. The possible mechanisms suggested are increase in binding of PPAR-y (Peroxisome ProliferatorActivated Receptor-gamma), inhibition of PTP-1B (Protein tyrosine phosphatase 1B), DPP-IV (Dipeptidyl peptidase IV), IX-amylase, IX-glucosidase, tissue glucose uptake and reduction in the oxidative stress (Unpublished data, See Fig 1). The pharmacodynamic complexity is fitly matched by the diversity of targets addressing euglycemia in the body. While restoring or correcting these anomalies individually, one at a time, is the objective of conventional antidiabetic therapy, herbal products appear to tackle many of these targets in a concerted manner, providing a more sustainable method with perhaps a lower incidence of adverse events (Veerapur, 2007). REVERSE PHARMACOLOGY: AN EFFECTIVE STRATEGY The beaten track of drug discovery rests on the paradigm that the course of innovation begins in the lab and ends in the clinic. Reverse pharmacology is a complete turnaround in strategy, where clinical evidence works at the root of drug discovery (Patwardhan et at., 2004). It is essentially a clinic to lab strategy, working backwards, where existing claims by traditional systems of medicine, such as Ayurveda, give clues to possible clinical value of existing formulations in traditional medicine. In such situations, clinical safety and proof of concept are the premises upon which the program is built, thus saving millions of dollars that are customarily spent on ensuring clinical safety and meeting regulatory rigours. Consequently, efficacy becomes a matter of validation, with the help of scientific evidence gathered from laboratory research. It is therefore a confluence of traditional wisdom, modern medicine and science. If exploited intelligently, it becomes a viable mode of drug discovery where holistic systems and traditional wisdom are integrated with laboratory evidence and modern science. The major advantage of reverse pharmacology is that it helps in reducing three major bottlenecks that plague the drug discovery programmes viz., time, money and toxicity. These bottlenecks are major hurdles in drug discovery impeding not only the pace of its progress but also increasing the costs. Increase in regulatory rigour has become one of the major reasons for the ever diminishing entry of NCEs. In this context, reverse pharmacology appears to be a viable alternate strategy. Several industries have realized the need for reinventing the drug discovery strategies for innovating with value added products.
Ayurveda and such other traditional systems of alternate medicine have a rich knowledge base with a vast materia medica consisting of thousands of plants, minerals, animal products used singly or in combinations, with various processes for the formulation of medicines. Most
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
23
of these formulations and therapeutic strategies have survived on the basis of evidence that have sustained centuries of clinical experience. However, lack of standardization and validation continues to undermine the pharmacopoeias of alternate medicine. It is in this context that modern medicine and science can act as a complementary support for establishing proof of concept and validation of claims.
CONCLUSIONS The last three decades have witnessed an unprecedented worldwide growth in the interest in NP- based medicines, both in developed and developing countries. Historical evidence shows that NPs offer a resource-pool for lead identification in drug discovery. NPs have a reasonable likelihood of biological activity, and isolation of a novel molecule is easier than de novo synthesis, especially when the molecular structure of the compound is very complex. NP-derived molecules are amenable to semisynthetic modifications and derivatisation. Such compounds are also significantly more complex and novel than most synthetic compounds, in addition to being chirally pure. Chiral specificity is very much in tune with the physiology of the body, giving the drug candidates a far greater level of specificity than nonchiral molecules. On account of such advantages, pharmaceutical companies are opting for NP based drug-leads. At present there is limited data on safety, efficacy, and clinical trials to prescribe NPs in therapy. However, the NP market has been continually growing at an increased rate during the last 30 years, mainly as nutraceutical, dietary supplements, cosmoceuticals and a few of them as chemopreventives. NP based therapy should emphasise on well-controlled and randomized clinical trials to prove safety and efficacy. International regulatory guidelines have to be followed in order to harmonise the use ofNPs. In some countries, the newer strategies and guidelines have been tried for the NPs (Briggs, 2002; Barnes, 2003; Vaidya et at., 2007; Calapai, 2008). Reverse pharmacology, which is driven by clinical evidence drawn from ancient wisdom, rather than preclinical data, also offers a viable time and cost saving strategy. Emphasis should also be given to the production and genetic improvement (whenever possible) of medicinal plants and other NPs. Recombinant DNA technology offers opportunities to synthesise the cryptic molecules of nature in a simple environmental-friendly way. Further, the emphasis must shift towards discovering leads from unexplored realms of biodiversity (Harvey, 2000; Harvey, 2007). Alternative sources for natural products as also the techniques to save the extinction of biodiversity should be given due importance for sustaining the program (Kennedy, 2008). Finally, but not any less important, is the need for a more detailed legislation on intellectual property rights related to the marketing of NPs. All these strategies are urgently needed to avoid both business conflicts and territorial conflicts on biodiversity.
24
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
REFERENCES Adrio, J.L. and Demain, A.L. (2006). Genetic improvement of processes yielding microbial products. FEMS Microbiol Rev., 30(2): 187-214. Auer, M., Moore, J.K, Meyer-Almes, J.F., Guenther, R, Pope, J.P. and Stoeckli, A.K (1998). Fluorescence correlation spectroscopy: lead discovery by miniaturized HTS. Drug Discov. Today, 3(10): 457-465. Baker, D.D., Chu, M., Oza, U. and Rajgarhia, V. (2007). The value of natural products to future pharmaceutical discovery. Nat Prod Rep., 24(6): 1225-1244. Balk, E.M., Tatsioni, A., Lichtenstein, A.H., Lau, J. and Pittas, A.G. (2007). Effect of chromium supplementation on glucose metabolism and lipids: a systematic review of randomized controlled trials. Diabetes Care, 30(8): 2154-2163. Bao, B., Sun, Q., Yao, X., Hong, J., Lee, C.O., Cho, H.Y. and Jung, J.H. (2007). Bisindole Alkaloids of the Topsentin and Hamacanthin Classes from a Marine Sponge Spongosorites sp. J Nat Prod., 70(1): 2-8. Barnes, J. (2003). Pharmacovigilance of herbal medicines: a UK perspective. Drug Sat, 26(12): 829-85l. Beckmann, N., Kneuer, R, Gremlich, H.U., Karmouty-Quintana, H., Ble, F.x. and Miiller, M. (2007). In vivo mouse imaging and spectroscopy in drug discovery. NMR Biomed., 20(3): 154-185. Beger, RD. (2006). Computational modeling of biologically active molecules using NMR spectra. Drug Discov Today, 11(9-10): 429-435. Bentley, R (2005). The development of penicillin: genesis of a famous antibiotic. Perspect Biol Med., 48(3): 444-452. Beranek, J. (1986). A study on structure-activity relationships of nucleoside analogues. Drugs Exp Clin Res., 12(4): 355-367. Bergmann, W. and Feeney, RJ. (1950). The isolation of a new thymine pentoside from sponge. JAm Chem Soc., 72: 2809-2810. Bilska, A. and Wtodek, L. (2005). Lipoic acid - the drug of the future? Pharmacol Rep., 57(5): 570-577. Blunt, J.W., Copp, B.R, Hu, W.P., Munro, M.H., Northcote, P.T. and Prinsep, M.R. (2007). Marine natural products. Nat Prod Rep., 24(1): 31-86. Bobzin, S.C., Yang, S. and Kasten, T.P. (2000). LC-NMR: a new tool to expedite the dereplication and identification of natural products. J Ind Mlcrobiol Bwtechnol., 25(6): 342-345. Boldi, A.M. (2004). Libraries from natural product-like scaffolds. Curro Opin. Chem. Biol., 8(3): 281-286. Brana, M.F. and Sanchez-Miga1l6n, A. (2006). Anticancer drug discovery and pharmaceutical chemistry: a history. Clin Transl Oncol., 8(10): 717-728. Briggs, D.R (2002). The regulation of herbal medicines in Australia. Toxicology, 181-182: 565-570 Brown, A.G., Butterworth, D., Cole, M., Hanscomb, G., Hood, J.D., Reading, C. and Rolinson, G.N. (1976). Naturally-occurring beta-lactamase inhibitors with antibacterial activity. J Antibiot (Tokyo)., 29(6): 668-669. Bulger, P.G., Bagal, S.K and Marquez, R (2008). Recent advances in biomimetic natural product synthesis. Nat Prod Rep., 25(2): 254-297. Burke, T.J., Loniello, KR, Beebe, J.A. and Ervin, KM. (2003). Development and application of fluorescence polarization assays in drug discovery. Comb Chem High Throughput Screen, 6(3): 183-194. Butler, M.S. (2004). The role of natural product chemistry in drug discovery. J Nat Prod., 67(12): 2141-2153. Butler, M.S. (2008). Natural products to drugs: natural product-derived compounds in clinical trials. Nat Prod Rep., 25(3): 475-516. Butler, M.S. and Newman, D.J. (2008). Mother Nature's gifts to diseases of man: the impact of natural products on anti-infective, anticholestemics and anticancer drug discovery. Prog Drug Res., 65(1): 3-44.
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
25
Calapai, G. (2008). European legislation on herbal medicines: a look into the future. Drug Sat, 31(5): 428-43l. Castel, D., Pitaval, A., Debily, M.A. and Gidrol, X. (2006). Cell micro arrays in drug discovery. Drug Discov Today, 11(13-14): 616-622. Castro-Perez, J.M. (2007). Current and future trends in the application of HPLC-MS to metabolite-identification studies. Drug Discov Today, 12(5-6): 249-256. Chaturvedi, H.C., Jain, M. and Kidwai, N.R (2007). Cloning of medicinal plants through tissue culture - a review. Indian J Exp Biol., 45(11): 937-948. Chiasson, J.L. and Rabasa-Lhoret, R (2004). Prevention of type 2 diabetes: insulin resistance and beta-cell function. Diabetes, 53(13): S34-S38. Chin, Y.W., Balunas, M.J., Chai, H.B. and Kinghorn, A.D. (2006). Drug discovery from natural sources. AAPS Journal, 8(2): E240-E253. Colburn, W.A. (2003). Biomarkers in drug discovery and development: from target identification through drug marketing. J. Clin. Pharmacol., 43: 329-34l. Coles, M., Heller, M. and Kessler, H. (2003). NMR-based screening technologies. Drug Discov. Today, 8(17): 803-810. Corcoran, O. and Spraul, M. (2003), LC-NMR-MS in drug discovery. Drug Discov. Today, 8(14): 624-63l. Cragg, G.M., Newman, D.J. and Yang, S.S. (2006). Natural product extracts of plant and marine origin having antileukemia potential. The NCI experience. J Nat Prod., 69(3): 488-498. Dagogo-Jack, S. (2006). Primary prevention of type-2 diabetes in developing countries. J Natl Med Assoc., 98(3): 415-419. Dickson, M. and Gagnon, J.P. (2004). Key factors in the rising cost of new drug discovery and development. Nat Rev Drug Discov., 3(5): 417-429. Diers, J.A., Ivey, K.D., EI-Alfy, A., Shaikh, J., Wang, J., Kochanowska, A.J., Stoker, J.F., Hamann, M.T. and Matsumoto, RR (2008). Identification of antidepressant drug leads through the evaluation of marine natural products with neuropsychiatric pharmacophores. Pharmacol Biochem Behav., 89(1): 46-53. DiMasi, J.A., Hansen, RW. and Grabowski, H.G. (2003), The price of innovation: new estimates of drug development costs. J Health Econ., 22: 151-185. Donia, M. and Hamann, M.T. (2003). Marine natural products and their potential applications as anti-infective agents. Lancet Infect Dis., 3(6): 338-348. Dougherty, T.J. and Miller, P.F. (2006). Microbial genomics and drug discovery: exploring innovative routes of drug discovery in the postgenomic era. IDrugs, 9(6): 420-422. Eisenstein, E.L., Collins, R, Cracknell, B.S., Podesta, 0., Reid, E.D., Sandercock, P., Shakhov, Y., Terrin, M.L., Sellers, M.A., Califf, RM., Granger, C.B. and Diaz, R (2008). Sensible approaches for reducing clinical trial costs. Clin Tnals, 5(1): 75-84. EI-Gammal, S.Y. (1994). Ancient Egyptian roots in the modern medical and pharmaceutical civilisation. Bull Indian Inst Hist Med Hyderabad, 24(2): 120-126. Endo, A. (1981). 3-Hydroxy-3-methylglutaryl-CoA reductase inhibitors. Methods Enzymol., 72: 684-689. Feng, X. and Siegel, M.M. (2007). FTICR-MS applications for the structure determination of natural products. Anal Bwanal Chem., 389(5): 1341-1363. Frank, RG. (2003). New estimates of drug development costs. J Health Econ., 22(2): 325-330. Frey, E.F. (1986). The earliest medical texts. Clio Med., 20(1-4): 79-90. Ganesan, A. (2004). Natural products as a hunting ground for combinatorial chemistry. Curr Opin Biotechnol., 15: 584-590. Ghofrani, H.A., Osterloh, I.H. and Grimminger, F. (2006). Sildenal: from angina to erectile dysfunction to pulmonary hypertension and beyond. Nat Rev Drug Discov., 5(8): 689-702. GOmez-Galera, S., Pelacho, A.M., Gene, A., Capell, T. and Christou, P. (2007). The genetic manipulation of medicinal and aromatic plants. Plant Cell Rep., 26(10): 1689-1715. Gordaliza, M. (2007). Natural products as leads to anticancer drugs. Clin Transl Oncol., 9(12): 767-776. Graul, A.I. (2007). Promoting, improving and accelerating the drug development and approval processes. Drug News Perspect., 20(1): 45-55.
26
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation [
Griffin, J.H., Linsell, M.S., Nodwell, M.B., Chen, Q., Pace, J.L., Quast, KL., Krause, KM., Farrington, L., Wu, T.X., Higgins, D.L., Jenkins, T.E., Christensen, B.G. and Judice, J.K (2003). Multivalent drug design. Synthesis and in vitro analysis of an array of vancomycin dimers. J Am Chern Soc., 125(21): 6517-6531. Gullo, V.P. and Hughes, E.D. (2005). Exploiting new approaches for natural product drug discovery in the biotechnology industry. Drug Discov Today: Technologies, 2(3): 281286. Haefner, B. (2003). Drugs from the deep: marine natural products as drug candidates. Drug Discou Today, 8(12): 536-544. Harvey, A. (2000). Strategies for discovering drugs from previously unexplored natural products. Drug D~scou Today, 5(7): 294-300. Harvey, A.L. (2007). Natural products as a screening resource. Curr Opin Chern BioI., 11(5): 480-484. Hays, N.P., Galassetti, P.R. and Coker, R.H. (2008). Prevention and treatment of type 2 diabetes: Current role of lifestyle, natural product, and pharmacological interventions. Pharmacol Ther., 118(2): 181-191. Home, P. (2003). The challenge of poorly controlled diabetes mellitus. Diabetes Metab., 29(2 Pt 1): 101-109. Hutchins, G.D., Miller, M.A., Soon, V.C. and Receveur, T. (2008). Small animal PET imaging. [LAB J., 49(1): 54-65. Inglese, J., Johnson, R.L., Simeonov, A., Xia, M., Zheng, W., Austin, C.P. and Auld, D.S. (2007). High-throughput screening assays for the identification of chemical probes. Nat Chern BioI., 3(8): 466-479. Jenwitheesuk, E., Horst, J.A., Rivas, KL., VanVoorhis, W.C. and Samudrala, R. (2008). Novel paradigms for drug discovery: computational multitarget screening. Trends Pharmacol Sci., 29(2): 62-71. Jimeno, J., Lopez-Martin, J.A., Ruiz-Casado, A., Izquierdo, M.A., Scheuer, P.J. and Rinehart, K (2004). Progress in the clinical development of new marine-derived anticancer compounds. Anticancer Drugs, 15(4): 321-329. Kahan, B.D. (1999). Cyclosporine: a revolution in transplantation. Transplant Proc., 31(1-2A): 14S-15S. Kennedy, J. (2008). Mutasynthesis, chemobiosynthesis, and back to semi-synthesis: combining synthetic chemistry and biosynthetic engineering for diversifying natural products. Nat Prod Rep., 25(1): 25-34. Kissau, L., Stahl, P., Mazitschek, R., Giannis, A. and Waldmann, H. (2003). Development of natural product-derived receptor tyrosine kinase inhibitors based on conservation of protein domain fold. J Med Chern., 46(14): 2917-2931. Knight, V., Sanglier, J.J., DiTullio, D., Braccili, S., Bonner, P., Waters, J., Hughes, D. and Zhang, L. (2003). Diversifying microbial natural products for drug discovery. Appl Microbiol Biotechnol., 62(5-6): 446-458. Koehn, F.E. and Carter, G.T. (2005). The evolving role of natural products in drug discovery. Nat Reu Drug Discou., 4(3): 206-220. Koehn, F.E. (2008). High impact technologies for natural products screening. Prog Drug Res., 65(175): 177-210. Konishi, Y., Kiyota, T., Draghici, C., Gao, J.M., Yeboah, F., Acoca, S., Jarussophon, S. and Purisima, E. (2007). Molecular formula analysis by an MSIMSIMS technique to expedite dereplication of natural products. Anal Chern., 79(3): 1187-1197. Koppitz, M. and Eis, K (2006). Automated medicinal chemistry. Drug Discou. Today, 11(11-12): 561-568. Korfmacher, W.A. (2005). Principles and applications of LC-MS in new drug discovery. Drug Discou. Today, 10(20): 1357-1367. Ladame, S. (2008). Dynamic combinatorial chemistry: on the road to fulfilling the promise. Org Biomol Chern., 6(2): 219-226. Lawrence, S. (2005). Biotech drug market steadily expands. Nat Biotechnol., 23(12): 1466. Lawrence, S. (2007). Pipelines turn to biotech. Nat Biotechnol., 25(12): 1342. Lee, M.L. and Schneider, G. (2001). Scaffold architecture and pharmacophoric properties of natural products and trade drugs: application in the design of natural product-based combinatorial libraries. J Comb Chern., 3(3): 284-289.
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
27
Li, Q.Y., Zu, Y.G., Shi, R.Z. and Yao, L.P. (2006). Review camptothecin: current perspectives. Curr Med Chem., 13(17): 2021-2039. Liao, Y. (2003). Diversity oriented synthesis and branching reaction pathway to generate natural product-like compounds. Curr Med Chem., 10: 2285-2316. Ligon, B.L. (2004). Penicillin: its discovery and early development. Semin Pediatr Infect Dis., 15(1): 52-57. Liu, B., Li, S. and Hu, J. (2004). Technological advances in high-throughput screening. Am J Pharmacogenomics, 4(4): 263-276. Lombardo, Y.B. and Chicco, A.G. (2006). Effects of dietary polyunsaturated or3 fatty acids on dyslipidemia and insulin resistance in rodents and humans. A review. J Nutr Biochem., 17(1): 1-13. Ma, S., Chowdhury, S.K and Alton, KB. (2006). Application of mass spectrometry for metabolite identification. Curr Drug Metab., 7(5): 503-523. Manzoni, M. and Rollini, M. (2002). Biosynthesis and biotechnological production of statins by filamentous fungi and application of these cholesterol-lowering drugs. Appl Microbiol Biotechnol., 58(5): 555-564. Marcaurelle, L.A. and Johannes, C.W. (2008). Application of natural product-inspired diversity-oriented synthesis to drug discovery. Prog Drug Res., 66(187): 189-216. Marris, E. (2006). Marine natural products: drugs from the deep. Nature, 443(7114): 904-905. Matfin, G. (2008). Challenges in developing drugs for the metabolic syndrome. Curr Diab Rep., 8(1): 31-36. Maugh, T.H. 2nd (1983). A survey of separative techniques. Science, 222(4621): 259-266. McChesney, J.D., Venkataraman, S.K and Henri, J.T. (2007). Plant natural products: back to the future or into extinction? Phytochemistry, 68(14): 2015-2022. McMahon, J.B., Beutler, J.A., O'Keefe, B.R., Goodrum, C.B., Myers, M.A. and Boyd, M.R. (2000). Development of a cyanovirin-N-HIV-1 gp120 binding assay for high throughput screening of natural product extracts by time-resolved fluorescence. J Biomol Screen., 5(3): 169-176. Mishra, KP., Ganju, L., Sairam, M., Banerjee, P.K and Sawhney, R.C. (2008). A review of high throughput technology for the screening of natural products. Biomedicine & Pharmacotherapy, 62: 94-98. Modak, M., Dixit, P., Londhe, J., Ghaskadbi, S. and Devasagayam, T.P.A., (2007). Indian herbs and herbal drugs used for the treatment of diabetes. J Clin Biochem Nutr., 40: 163-173. Mukherjee, P.K, Maiti, K, Mukherjee, K and Houghton, KP. (2006). Leads from Indian medicinal plants with hypoglycemic potentials. J Ethnopharmacol., 106: 1-28. Munro, M.H., Blunt, J.W., Dumdei, E.J., Hickford, S.J., Lill, R.E., Li, S., Battershill, C.N. and Duckworth, A.R. (1999). The discovery and development of marine compounds with pharmaceutical potential. J Biotechnol., 70(1-3): 15-25. Newman, D.J., Cragg, G.M. and Snader, KM. (2003). Natural products as sources of new drugs over the period 1981-2002. J Nat Prod., 66(7): 1022-1037. Newman, D.J. and Cragg, G.M. (2004). Marine natural products and related compounds in clinical and advanced preclinical trials. J Nat Prod., 67(8): 1216-1238. Newman, D.J. and Cragg, G.M. (2007). Natural products as sources of new drugs over the last 25 years. J Nat Prod., 70(3): 461-77. Nielsen, J. (2002). Combinatorial synthesis of natural products. Curr Opin Chem Biol., 6(3): 297-305. O'Connell, D. and Roblin, D. (2006). Translational research in the pharmaceutical industry: from bench to bedside. Drug Discov Today, 11(17/18): 833-838. Ortholand, J.Y. and Ganesan, A. (2004). Natural products and combinatorial chemistry: back to the future. Curr Opin Chem Bwl., 8(3): 271-280. Oumeish, O.Y. (1998). The phillosophical, culture, and historical aspects of complementary, alternative, unconventional and integrative medicine in the old world. Arch Dermatol., 134: 1373-1386. Pal, S.K and Shukla, Y. (2003). Herbal medicine: current status and the future. Asian Pac J Cancer Prev., 4(4): 281-288.
28
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Patwardhan, B., Vaidya, D.B.A. and Chorghade, M. (2004). Ayurveda and natural products drug discovery. Curr Sci., 86(6): 789-799. Paul, V.J., Arthur, KE., Ritson-Williams, R., Ross, C. and Sharp, K (2007). Chemical defenses: from compounds to communities. Bwl Bull., 213(3): 226-251. Pauli, F.G., Jaki, U.B. and Lankin, C.D. (2005). Quantitative 1H NMR: Development and potential of a method for natural products analysis. J Nat Prod., 68: 133-149. Paulus, M.P. and Stein, M.B. (2007). Role of functional magnetic resonance imaging in drug discovery. Neuropsychol Rev., 17(2): 179-88. Pereira, D.A. and Williams, J.A (2007). Origin and evolution of high throughput screening. Br J Pharmacal., 152(1): 53-61. Phillipson, D.W., Milgram, KE., Yanovsky, AI., Rusnak, L.S., Haggerty, D.A., Farrell, W.P., Greig, M.J., Xiong, X. and Proefke, M.L. (2002). High-throughput bioassay-guided fractionation: a technique for rapidly assigning observed activity to individual components of combinatorial libraries, screened in HTS bioassays. J Comb Chem., 4(6): 591-599. Pillai, O. and Panchagnula, R. (2001). Insulin therapies - past, present and future. Drug Discov Today, 6(20): 1056-1061. Posner, B.A (2005). High-throughput screening-driven lead discovery: meeting the challenges of finding new therapeutics. Curr Opin Drug Discov. Devel., 8(4): 487-494. Potterat, O. and Hamburger, M. (2008). Drug discovery and development with plant-derived compounds. Prog Drug Res., 65(45): 47-118. Powers, R., Mercier, KA and Copeland, J.C. (2008). The application of FAST-NMR for the identification of novel drug discovery targets. Drug Discov. Today, 13(3-4): 172-179. Qiu, J. (2007). 'Back to the future' for Chinese herbal medicines. Nat Rev Drug Discov., 6(7): 506-507. Rawat, D.S., Joshi, M.C., Joshi, P. and Atheaya, H. (2006). Marine peptides and related compounds in clinical trial. Anticancer Agents Med Chem., 6(1): 33-40. Reid, D.G. and Murphy, P.S. (2008). Fluorine magnetic resonance in vivo: A powerful tool in the study of drug distribution and metabolism. Drug Discov. Today, 13(11-12): 473-480. Robert, C.K.G. (2000). Applications of NMR in drug discovery. Drug Discov. Today, 5(6): 230-240. Robert-II, J.L. and Morrow, D.J. (2001). In: Goodman and Gilman's Pharmacological basis of therapeutics. 10th Edn; by Hardman, G.J., Limbird E.L, McGraw Hill international edition, Chapter 27, pp 687-688. Rogers, M.V. (1997). Light on high-throughput screening: fluorescence-based assay technologies. Drug Discov. Today, 2(4): 156-160. Ross, A. and Senn, H. (2001). Automation of measurements and data evaluation in biomolecular NMR screening. Drug Discov. Today, 6(11): 583-593. Rouhi, AM. (2003). Moving beyond natural products. Chem Eng News, 81: 104-107. Saklani, A and Kutty, S.K (2008). Plant-derived compounds in clinical trials. Drug Discov. Today, 13(3-4): 161-171. Saleem, M., Ali, M.S., Hussain, S., Jabbar, A, Ashraf, M. and Lee, Y.S. (2007). Marine natural products offungal origin. Nat Prod Rep., 24(5): 1142-1152. Sartor, R.B. (2003). Translational research: Bridging the widening gap between basic and clinical research. Gastroenterology, 124(5): 1178. Saxena, A and Vikram, N.K (2004). Role of selected Indian plants in management oftype 2 diabetes: a review. J Altern Complement Med., 10(2): 369-378. Schmid, E.F. and Smith, D.A. (2007). Pharmaceutical R&D in the spotlight: why is there still unmet medical need? Drug Discov. Today, 12(23-24): 998-1006. Schuffenhauer, A, Ruedisser, S., Marzinzik, A.L., Jahnke, W., Blommers, M., Selzer, P. and Jacoby, E. (2005). Library design for fragment based screening. Curr Top Med Chem., 5(8): 751-762. Senior, K. (2005). Wide-angle X-ray scattering for screening functional ligands. Drug Discov. Today, 10(3): 163. Shang, S. and Tan, D.S. (2005). Advancing chemistry and biology through diversityoriented synthesis of natural product-like libraries. Curr Opin Chem Biol., 9(3): 248258.
Efficacy, Safety and Clinical Evaluation of Bioactive Natural Products
29
Sonntag, KC. (2005). Implementations of translational medicine. J Transl Med., 3: 33. Starkuviene, V. and Pepperkok, R. (2007). The potential of high-content high-throughput microscopy in drug discovery. Br J Pharmacol., 152: 62-71. Strege, M.A. (1999). High-performance liquid chromatographic-electro spray ionization mass spectrometric analyses for the integration of natural products with modern high-throughput screening. J Chromatogr B Biomed Sci Appl., 725(1): 67-78. Suckling, C. (1991). Chemical approaches to the discovery of new drugs. Sci Prog., 75(298 Pt 3-4): 323-359. Sultana, R.S., Roblin, D. and O'Connell, D. (2007), Translational research in the pharmaceutical industry: from theory to reality. Drug Discov Today, 12(9/10): 418425. Tan, D.S. (2004). Current progress in natural product-like libraries for discovery screening. Comb Chem High Throughput Screen., 7(7): 631-643. Tan, D.S. (2005). Diversity-oriented synthesis: exploring the intersections between chemistry and biology. Nat Chem BioI., 1: 74-84. Tan, L.T. (2007). Bioactive natural products from marine cyanobacteria for drug discovery. Phytochemistry, 68(7): 954-979. Trusheim, M.R., Berndt, E.R. and Douglas, F.L. (2007). Stratified medicine: strategic and economic implications of combining drugs and clinical biomarkers. Nat Reu Drug Discou., 6(4): 287-293. Uemura, D. (2006). Bioorganic studies on marine natural products-diverse chemical structures and bioactivities. Chem Rec., 6(5): 235-248. Vaidya, A.D. and Devasagayam, T.P. (2007). Current status of herbal drugs in India: an overview. J Clm Biochem Nutr., 41: 1-11. Vane, J.R. (2000). The fight against rheumatism: from willow bark to COX-l sparing drugs. J Physiol Pharmacol., 51(4 Pt 1): 573-86. Veerapur, V.P. (2007). Activity guided phytopharmacological analysis of selected Indian medicinal plants with special reference to diabetes mellitus. PhD thesis, submitted to Manipal University, India, for the award of doctoral degree. Wainwright, M. (1990). Miracle Cure: The Story of Penicillin and the Golden Age of Antibiotics; Blackwell: Oxford, UK Wang, J. and Maurer, L. (2005). Positron Emission Tomography: applications in drug discovery and drug development. Curr Top Med Chem., 5(11): 1053-1075. Wehling, M. (2006). Translational medicine: can it really facilitate the transition ofresearch "from bench to bedside"? Eur J Clin Pharmacol., 62: 91-95. Wild, S., Roglic, G., Green, A., Sicree, R. and King, H. (2004). Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care, 27(5): 1047-1053. Wild, S., Roglic, G., Green, A., Sicree, R. and King, H. (2007). Screening for type 2 diabetes: literature review and economic modeling. Health Technol Assess., 11(17): iiiiv, ix-xi, 1-125. Wilkinson, B. and Moss, S.J. (2005). Biosynthetic engineering of natural products for lead optimization and development. Curr Opin Drug Dlscov. Deuel., 8(6): 748-756. Xie, J.T., Mchendale, S. and Yuan, C.S. (2005). Ginseng and diabetes. Am J Chin Med., 33(3): 397-404. Xing, J., Xie, C. and Lou, H. (2007). Recent applications of liquid chromatography-mass spectrometry in natural products bioanalysis. J Pharm Biomed Anal., 44(2): 368-378. Zemanova, L., Schenk, A., ValIer, M.J., Nienhaus, G.U. and Heilker, R. (2003). Confocal optics microscopy for biochemical and cellular high-throughput screening. Drug Discov. Today, 8(23): 1085-1093. Zhang, J., McCombie, G., Guenat, C. and Knochenmuss, R. (2005). FT-ICR mass spectrometry in the drug discovery process. Drug Discou. Today, 10(9): 635-642. Zhang, Y., Wu, D.R, Wang-Iverson, D.B. and Tymiak, A.A. (2005). Enantioselective chromatography in drug discovery. Drug Discou. Today, 10(8): 571-577. Zheng, F. and Zhan, C.G. (2008). Structure-and-mechanism-based design and discovery of therapeutics for cocaine overdose and addiction. Org Biomol Chem., 6(5): 836-843.
"This page is Intentionally Left Blank"
2 The Modern Application of Traditional Chinese Medicine: Indigo N aturalis as an Alternative Therapy in Psoriasis vulgaris YIN-Ku LIN 1 ,2 AND JONG-HWEI Su PANG2 ,*
ABSTRACT
Psoriasis is a distressing, recurrent disease that significantly impairs the quality of life and has no permanent cure. Traditional Chinese medicine is an alternative method of therapy that can be used in the psoriasis treatment. It has been reported in Chinese literature that indigo naturalis exhibited potential anti-psoriatic effects in systemic therapy. However, its pharmacology was unclear and its gastrointestinal side effects limited its use in clinical practice. This article reviews and summarizes our experience of psoriasis therapy using Chinese herb prescription and the investigation of the anti-psoriatic mechanism of indigo naturalis. Indigo naturalis formulated in ointment has efficacy in treating psoriasis and can avoid adverse systemic effects. The possible mechanism of indigo naturalis action in treating psoriasis includes inhibiting hyperproliferation of epidermal keratinocytes and lymphocytes infiltration, as well as restoring epidermal differentiation and tight junctions in psoriatic skin. We believe that indigo naturalis will become a promising natural product that exerts great therapeutic potential as an alternative therapy in psonas£s. Key words: Traditional Chinese medicine, psoriasis, indigo naturalis, indirubin, keratinocyte, differentiation 1. Department of Traditional Chinese Medicine, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, 204, Taiwan. 2. Graduate Institute of Clinical Medical Sciences, Chang Gung University, Tao-Yuan, 333, Taiwan. * Corresponding author: E-mail:
[email protected]
32
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
INTRODUCTION Psoriasis is a chronic inflammatory disease of the skin characterized by generalized erythematous papules or well-demarcated erythematous plaques topped by silvery scales which vary in severity and clinical manifestation. The most common form, plaque psoriasis, affects 2-3% of the Caucasian population (Lebwohl, 2003) but only 0.3% of the Mongoloid population (Yip, 1984). Many factors such as genetic features, immunological disorders, biochemical changes, environmental effects, trauma, stress, endocrine features, infection, drugs and alcohol play important roles in the pathogenesis of psoriasis (Ortonne, 1996). Conventional treatment for psoriasis has several disadvantages, including disease rebound or worsening after treatment is stopped (e.g. steroid) and risk for organ toxicities, such as nephrotoxicity and hepatotoxicity (e.g. cyclosporine, methotrexate). The fear of adverse events in long-term use may lead to poor compliance and results in following flares. Therefore, it is important to explore an alternatively new intervention which is relatively safe and can prolong the remission duration of psoriasis or prevent progression. The earliest description of psoriasis in ancient Chinese medical literature may be found in the 5th century AD. Traditionally, Chinese herbal preparation has been extensively used to treat psoriasis both topically and orally for centuries, and produced promising clinical results (Bedi & Shenefelt, 2002). Most of the key actions of Chinese herbs commonly used in treating psoriasis reflect anti-inflammatory properties, modulation of cytokine production or inhibition of angiogenesis (Tse, 2003). All these actions are potentially relevant in reducing the severity of psoriasis. Nowadays, traditional Chinese medicine (TCM) is still one of the most frequently chosen alternative therapies that can be used in the treatment of psoriasis in China and Taiwan. However, its lack of randomized blinded control trials and its underlying mechanisms of action have not been systemically investigated (Koo & Desai, 2003). The theoretical system and the terminology ofTCM are quite different from those of Western medicine. The doctors examine their patients with four traditional diagnostic methods (inspection, auscultation, interrogation, and pulse-taking), determine the principle of treatment according to the differentiation of symptoms and signs, and prescribe a formula according to the principle of treatment. In general, standard TCM practice emphasizes the importance of using many herbs that are combined in different formulations for each individual patient, sometimes mixing more than 10 different herbs. The mixture of herbs prescribed varies individually depending on the subtype of psoriasis (''blood-heat'' type, "blood deficiency dryness" type, and ''blood stasis" type), which is determined in traditional Chinese by many findings, including the lesions of psoriasis, the pulse, and the condition of the tongue (Tse, 2003). This is vastly different from Western medicine, which uses therapeutic approaches that are standardized and mono-therapy, as well as emphasizing
Indigo Naturalis as an Alternative Therapy in Psoriasis vulgaris
33
"average" efficacy in large, double-blinded, placebo-controlled studies. Due to the overwhelming variability in how patients are treated, both between TCM practitioners and within the practice of a single TCM practitioner, it is difficult to conduct controlled trials to study traditional approach of individual polypharmacy. Indigo naturalis is one of the Chinese herbal remedies that have been reported to exhibit potential anti-psoriatic efficacy (Yuan et a!., 1982). However, the poor solubility and low absorption of indigo naturalis, and the adverse effects associated with irritation of the gastrointestinal tract and liver function in long-term systemic use have been occasionally reported (Verucchi et al., 2002). In order to avoid the adverse systemic effects, but retain the demonstrated efficacy of indigo naturalis as an anti-psoriatic medicine, we initiated an alternative approach in 2003 by applying the drug topically on skin lesions. Within the last five years, we have been using indigo naturalis formulated in ointment topically to treat thousands of patients with psoriasis. We also used indigo naturalis as a mono-therapy in treating psoriasis and conducted randomized, observer-blinded, vehicle controlled, intra-patient comparison trials to evaluate the clinical efficacy and safety in patients with psoriasis treated with indigo naturalis. This unique mono-therapy, rather than individual polypharmacy that TCM used to emphasize, hence provided a compelling evidence of indigo naturalis for treating psoriasis. This review aims to provide our clinical experience for the efficacy, safety of indigo naturalis in topical treatment of psoriasis, and our studies on the pharmacology of indigo naturalis and its active components.
INDIGO NATURALlS'S PROFILE Indigo naturalis (Qing Dai) is a dark blue powder originating from the branches and leaves of various indigo-producing plants, such as Strobilanthes fiaccidifolius, Polygonum tinctorium, Baphicacanthus cusia Bremek, and Isatis indigotica Fort. The extraction process is based on the natural fermentation method in Taiwan. In brief, the freshly cut plants are soaked in water until they decompose and the contents of indigo naturalis are ready to be extracted. An adequate amount of limestone powder is then added and oxygen is forced into the liquid usually by stirring the soaking solution thoroughly until it turns from blackish green to deep red, extracted products contained in the foaming surface of the mixture are collected and air-dried for later use. Indigo naturalis was used as textile dyes, paint pigment and medicine in ancient China. Indigo naturalis and indigo concoctions are recorded as being applied externally or taken internally before the first century AD in China and India. Until the mid-twentieth century, when antibiotics and steroids came into the world, indigo naturalis was one of the most important
34
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
and efficacious treatments to heal ailments of various infections and inflammatory diseases in China and Taiwan. There are many people today who still have faith in the powers of indigo naturalis to cure various difficult illnesses, such as cancer. In TCM, indigo naturalis has been described as possessing antiinflammatory, anti-microbial, analgesic and sedative properties, i.e. "clearing away heat, detoxicating, removing blood-heat and ecchymoses, purging liver-fire and arresting convulsion". Indigo naturalis has been used as a remedy for high fever in infectious sickness, infantile convulsions, epilepsy, aphtha, mumps, pharyngitis, tonsillitis, gingivitis, periodontitis, and various dermatoses, including eczema, eruption, erysipelas, carbuncles, furuncles and insect or snake bites. Modern research, particularly in China, is revealing that indigo naturalis, with effects of antivirus, and anti-bacteria, has also been used as an important source of anti-cancer drugs in China (Han, 1994). A well-studied example is Danggui Longhui Wan, a mixture of 11 herbal medicines traditionally utilized against certain types of leukemia. Only one of these ingredients, indigo naturalis, was found to carry the antileukemic activity (Xiao et al., 2002). Although indigo naturalis is mostly constituted of indigo blue, a minor constituent, indirubin, was identified as the active component (Hoessel et al., 1999).
TOPICALLY INDIGO NATURALIS IN PATIENTS WITH PSORIASIS Clinical Efficacy Since 2003, we began to use Chinese herbs topically to treat patients with psoriasis. Initially, we used a combination of three herbs, indigo naturalis, Scutellaria baicalensis-Georgi, and Cortes phellodendri, which was recommended for psoriatic treatment in TCM. The topical ointment combined mixed herbs and vaseline, yellow wax as well as olive oil. We found that many patients with psoriasis, after multiple treatment failures with conventional anti-psoriasis medications, showed clinical improvement after topical treatment with indigo naturalis composite ointment. In addition, remission time was found to last longer than their previous therapies. This novel finding was reported in the journal Pediatric Dermatology (Lin et al., 2006). An 8-year-old boy with widespread psoriasis showed remarkable improvement with 8 weeks of topical treatment with this ointment. The psoriatic lesions on his body disappeared completely and the total body surface area involvement (BSA) decreased from almost 80% to 0%. Remission has lasted for over 2 years. In past 6 years observation, only mild disease recurred with no more than 1% ofBSA was noticed in this patient which was immediately improved after re-treatment with indigo naturalis composite ointment. Because this formula consists of three herbs, it is difficult to conduct controlled trials and investigations of pharmacological mechanism. We reviewed the published literature, and
Indigo Naturalis as an Alternative Therapy in Psoriasis vulgaris
35
found indigo naturalis and its active ingredients, indirubin, have been orally used to treat psoriasis with considerable effectiveness (Koo & Arain, 1998). Therefore, we initiated the indigo naturalis mono-therapy for psoriasis and we reported that two adult patients with severe, recalcitrant psoriasis were successfully treated with topical indigo naturalis in the journal Clinical and Experimental Dermatology (Lin et al., 2006). Two patients were male aged 45 and 36. One had an 8-year history of psoriasis and was presented with 40% BSA and a Psoriasis Area and Severity Index (PAS!) score of 28.8. The other patient had suffered psoriasis for 22 years. He was presented with more than 80% BSA and a PASI score in excess of 42. The treatment by indigo naturalis ointment resulted in an approximately 95% reduction in PASI scores and BSA in these two patients within 3 months (Fig 1).
Fig 1. Clinical improvement after the treatment with indigo naturalis. A 40-year-old man with a 22-year history of plaques-type psoriasis . (A) Severe plaques are salmon pink in color, covered by silvery scales on right arm at the initial visit; (B) the same arm after 12 weeks of treatment with indigo naturalis ointment
In order to provide evidence-based data for this alternative therapy in psoriasis, we designed a randomized, vehicle-controlled clinical trial to evaluate the efficacy and safety of topically applied indigo naturalis in treating plaque-type psoriasis. An 8-week pilot clinical trial was performed during April-June 2004 in our hospital. Fourteen patients (10 men and 4 women) with chronic plaque psoriasis were enrolled. The mean patient age was 35.8 years (range, 21-54 years) and the median disease duration
36
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
was 10.8 years (range, 2-30 years). The median pretreatment PASI was 11.7, with a median BSA of 13.6%. The patients were topically treated with either indigo naturalis ointment or vehicle ointment on contralateral skin lesions daily for 8 weeks. Efficacy was evaluated on the basis ofthe clinical scores, including induration, scaling, erythema and clearing percentage. Ten patients completed the trial and the results demonstrated a significant reduction in clinical scores achieved by topically applied indigo naturalis ointment. This interesting result has been published in the journal Dermatology (Lin et al. , 2007). To further confirm the efficacy of indigo naturalis for psoriasis treatment, another randomized, observer-blinded, vehicle controlled, intra-patient comparative trial was conducted. Fortytwo (32 men, 10 women) outpatients with chronic plaque psoriasis were enrolled. The mean patient age was 34.6 years (range, 18-58 years) and the median disease duration was 10 years (range, 2-41 years). The median pretreatment PASI was 14.7, with a median BSA of 18%. Target lesions were located on the upper extremities (17 participants), lower extremities (21 participants), and trunk (4 participants). The patients applied either indigo naturalis ointment or vehicle ointment topically to each of two bilaterally symmetrical psoriatic plaque lesions for 12 weeks. There was no significant difference in score of area, redness, thickness, and scaling between lesions which were assigned for treatment by indigo naturalis and vehicle ointments in baseline. The outcomes were assessed with the sum of erythema, scaling, and in duration scores and the clearing percentage of the target plaque lesion by two blinded professional observers. A significant reduction in the sum of scaling, erythema and in duration scores (p
Indigo Naturalis as an Alternative Therapy in Psoriasis vulgaris
37
other currently used topical medicine (e.g. corticosteroid) to further evaluate the response duration and the disease recurrence. Histological Response to Indigo Naturalis Treatment in Psoriatic Lesions Based on our histological examination of skin biopsies, we found a marked decrease in epidermal thickness and elongated rete ridges for the lesions treated with indigo naturalis compared with the untreated lesions (Fig 2). We suggested that the anti-psoriatic effect of indigo naturalis might be mediated by controlling hyperproliferation and/or abnormal differentiation of epidermal keratinocytes, and/or exhibiting immunomodulatory effects in psoriatic lesion. We analyzed and compared the expression of different marker proteins with respect to proliferation (Ki-67, PCNA), differentiation (involucrin, filaggrin), and inflammation (CD3) in psoriatic lesions treated with indigo naturalis ointment and vehicle ointment. Results from immunohistochemical analysis showed a marked decrease in the number of cells with positively Ki-67 -stained nuclei in the indigo naturalis ointment-treated lesions compared to that in vehicle ointment-treated lesions (Lin et al., 2007). Similar findings were also demontrated for PCNA marker (Lin et al., 2007). The increased and normalized expression of filaggrin was observed in the stratum coreum and granular layer of the skin lesions treated with the indigo naturalis ointment (Lin et al., 2007). Involucrin also expressed normally in the granular and upper spinous layers after indigo naturalis therapy (Lin et al., 2009). In contrast to the original psoriatic lesions, the number of positively CD-3-stained T lymphocytes was demonstrated to be significantly reduced in the epidermis and papillary dermis of indigo naturalis ointment-treated lesions (Lin et al., 2007). In addition, based on our histological examination of skin biopsies, we found the presence of wide intercellular clefts only in the epidermis of lesions before treatment and control lesions treated with the vehicle ointment, but not in lesions treated with indigo naturalis ointment. In the treated psoriatic lesions that showed good clinical improvement, the keratinocytes in the granular layers were found to adhere to each other tightly. This finding implied that the impaired epidermal tight junction of psoriatic lesions may recover as a result of indigo naturalis treatment. We analyzed the expression of different marker proteins with respect to tight junction function (such as daudin-I) in psoriatic lesions, and the results showed a significant increase and recovery after treatment with topical indigo naturalis ointment compared with vehicle ointment (manuscript submitted).
38
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Fig 2. Effect of topically applied indigo naturalis ointment on the histological change of psoriatic lesion. Skin biopsies taken at baseline and after 8 weeks of treatment were processed for histological examination by HE stain method. (A) Untreated psoriatic lesion. (B) Indigo naturalis-treated lesion. Original magnification: xlOO
In short, our pathological findings suggested that indigo naturalis has great potential values in the treatment of psoriasis by inhibiting hyperproliferation of epidermal keratinocytes and lymphocytes infiltration, as well as restoring epidermal differentiation and tight junctions in psoriatic skin. Whether these findings are simply a result of skin recovered from psoriasis after indigo naturalis treatment or indigo naturalis might directly modulate these changes in skin is waited to be clarified by investigating the pharmacology of indigo naturalis in much more details.
Adverse Events Indigo naturalis, as a crude extract, appears to be dark blue in color and has a slightly unpleasant odor. The indigo naturalis ointment might slightly
Indigo Naturalis as an Alternative Therapy in Psoriasis vulgaris
39
stain the skin and clothing which can be cleaned thoroughly by common detergents. Repeated application has no significant effect on the skin color and does not change the skin appearance. However, either the color or smell of indigo naturalis may affect the compliance offew patients, therefore benefit comes along with longer treatment can not possibly attain for them. Among 10 patients who completed an 8-week pilot clinical trial, 2 patients reported a short period (3-5 days) of itching at the beginning of treatment. In the following treatment period, no further adverse events such as scaling, erythema, tenderness, vesicles or other serious problems were found (Lin et al., 2007). Another 12 weeks clinical trial, 4 of 34 patients who completed the treatment reported itching after applying the indigo naturalis ointment to their psoriatic plaque, but the itching was only present for a couple of days at the start of treatment, and no tenderness, vesicles or other serious problems were found in the subsequent treatment period (Lin et al., 2008). In these two study groups, there were no abnormalities seen in the liver function tests, renal function tests, and complete blood counts after trial. Similar results were obtained in our long-term follow-up of these patients for the past 5 years. In our past 5-years experience in clinic, there were no serious events such as hepatic toxicity, renal damage and gastrointestinal problems found in these studies. Hypersensitivity, however, may be the major adverse effect as we have noticed in clinic (less than 15%). Some patients developed erythrodema and itching after using topical indigo naturalis treatment for psoriasis and most of them could recover by discontinuing the indigo naturalis application. Combining the treatment with topical steroids intermittently the development of allergy contact dermatitis may also be avoided or diminished.
PHARMACOLOGICAL PROPERTIES OF INDIGO NATURALIS Although the effectiveness of indigo naturalis in the treatment of psoriasis is well established, the mechanism of its action is poorly understood. We have utilized cultured normal human epidermal keratinocytes (NHEKs) as a psoriasis-relevant experimental model to investigate the potential effects of indigo naturalis on the gene expression of keratinocytes. In our laboratory data, we found that indigo naturalis could suppress proliferation and promote differentiation of epidermal keratinocytes which were correlated with the down-rgulation of PCNA gene expression and the upregulation of involucrin gene expression of NHEKs (Lin et al., 2009). We have also made another novel finding that indigo naturalis can improve skin barrier protection, at least in part, by up-regulating claudin-1 gene expression and therefore the tight junction function (manuscript submitted). The main components of indigo naturalis used in our clinic are indigo blue, indirubin and tryptanthrin as identified in chromatographic fingerprints. Among them, indigo blue in the most abundant one, however,
40
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
the activity in psoriasis treatment has not been reported yet (Lin, 1993). Our preliminary data suggest that indigo blue may exert an antiangiogenic effect which may also partially explain the efficacy of indigo naturalis. Indigo blue extracted from some of the indigo-producing plants was reported to possess a hepatoprotective effect (Sreepriya et al., 2001). Indirubin and its analogues have been used to treat psoriasis orally in China (Lin, 1993). In addition, clinical trials showed that indirubin has a definite efficiency against chronic myelogenous leukemia (Han et al., 1988; Xiao et al., 2002). Indirubin and its analogues selectively inhibit CDKs and other kinases and block cell proliferation in the late G 1 and G2/M phase of the cell cycle (Marko et al., 2001). The promotion of neutrophilic differentiation of human myelocytic leukemia HL-60 cells by indirubin has also been reported (Suzuki et al., 2005). Furthermore, recent studies have revealed that indirubin simultaneously targets several signaling/molecules involved in the pathological process of cancer. The targets include those involved with cell cycle regulation (Hoessel et al., 1999), growth factor receptor signaling (Stat3) (Nam et al., 2005) and AhR signaling (Knockaert et al., 2004). However, mechanism underlying the efficacy of indirubin in psoriasis is still unclear, and the pharmacology of indirubin in keratinocytes also is unknown. Tryptanthrin, another bioactive ingredient from indigo-producing plants, has been reported to exhibit anti-tumor, anti-inflammatory and antiallergic activities (Lin, 1993). Whether tryptanthrin play any role in the anti-psoriatic effect of indigo naturalis is not elucidated and we will further investigate its pharmacology. SUMMARY
Although the western medicines currently used for psoriasis treatments can produce clinical improvement at controlling the disease, none are universally safe and effective, and each carries a considerable risk profile. The clinical responses are sometimes typically short duration, and may be accompanied by disease rebound after therapy is withdrawn. All these therapies for psoriasis are only remissive, but not curative. Recently, alternative therapy (such as herbal remedies, nutritional supplements, and dietary manipulation) for the treatment of psoriasis has increasingly received much attention (Traub & Marshall, 2007). Most common reason cited for patients with psoriasis who are interested in alternative therapy was the dissatisfaction with their conventional treatment. Chinese herbal medicines have demonstrated their efficacy in treating psoriasis for hundreds of years. Unfortunately, the use of traditional Chinese herbal medicine, a mixture of various herbs individually formulated for the patient makes scientific analysis extremely difficult. The mechanisms of these herbs were unknown and any possible adverse effect associated these herbs has fallen on dead ears. In this article, we attempt to provide evidence-based information on the clinical benefit of using topical indigo naturalis as a
Indigo Naturalis as an Alternative Therapy in Psoriasis vulgaris
41
mono-therapy for psoriasis and explore the potential mechanisms of indigo naturalis in treating psoriasis. In our experience, the use of topical application can avoid first-pass effect and chemical degradation of the drug in the gastrointestinal tract, as well as minimizing the risk of toxic side effects including gastric irritation, and liver damage. As psoriasis is a persistent and recurring disease, patients often require continuous treatment preferably with a low cumulative toxicity potential. The use of topical application not only presents direct and better therapeutic efficacy but also avoids the long-term adverse reactions. The effects for the use of indigo naturalis have been described in Chinese medical terms as "clearing the heat and cooling the blood" (Lin, 1993). However, the mechanisms underlying the effects of indigo naturalis specially for treating psoriasis seem to be very limited in the published literature. Psoriasis has three principal histological features: epidermal hyperplasia; dilated, prominent blood vessels in the dermis; and an inflammatory infiltrate of leucocytes, predominantly into the dermis (Griffiths & Barker, 2007). Since psoriatic plaques are likely to arise from the failure of normal differentiation process, reagents that suppress hyperproliferation and promote differentiation of epidermal keratinocytes, as well as inhibit inflammation may have a significant impact on the treatment of psoriasis. Our pathological findings verified the above hypothesis that indigo naturalis is indeed effective to improve psoriatic lesion by controlling hyperproliferation and abnormal differentiation of epidermal keratinocytes, as well as lymphocyte infiltration. In addition, as shown in our histological findings, indigo naturalis may have the effect of recovering the epidermal barrier function. In our in vitro study, we were able to demonstrate that the proliferation of epidermal keratinocytes was down-regulated and differentiation was up-regulated after indigo naturalis treatment. Further investigation of pharmacological mechanism of indigo naturalis and its active components in treating psoriasis are currently performed in our laboratory. In general, topical use of indigo naturalis is relatively safe compared to other currently used therapies. However, one should be aware that most ofthe herb indigo naturalis sold in the Taiwan area (this can happen in any area) is not prepared from natural plant. The chemically synthesized indigo naturalis barely contains any effective ingredient, stains the skin and clothing more easily, and tends to induce hypersensitivity. In conclusion, indigo naturalis topically used for treating psoriasis is both safe and effective. We are very pleased to share our experience in the clinic and basic studies of psoriasis treatment which may contribute to the development of promising therapeutic agents for psoriasis.
42
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
ACKNOWLEDGEMENTS Our studies were supported by Grants CMRPG33024 and CMRPG350301 from Chang Gung Memorial Hospital, Taiwan.
REFERENCES Bedi, M.K. and Shenefelt, P.D. (2002). Herbal therapy in dermatology. Archives of Dermatology, 138: 232-242. Griffiths, C.E. and Barker, J.N. (2007). Pathogenesis and clinical features of psoriasis. Lancet, 370: 263-271. Han, J. (1988). Traditional Chinese medicine and the search for new antineoplastic drugs. Journal of Ethnopharmacology, 24: 1-17. Han, R. (1994). Highlight on the studies of anticancer drugs derived from plants in China. Stem cells, 12: 53-63. Hoessel, R., Leclerc, S., Endicott, J.A., Nobel, M.E., Lawrie, A., Tunnah, P., Leost M, Damiens, E., Marie, D., Marko, D., Niederberger, E., Tang, W., Eisenbrand, G. and Meijer, L. (1999). Indirubin, the active constituent of a Chinese antileukaemia medicine, inhibits cyclindependent kinases. Nature Cell Biology, 1: 60-67. Knockaert, M., Blondel, M., Bach, S., Leost, M., Elbi, C. and Hager, G.L. (2004). Independent actions on cyclin-dependent kinases and aryl hydrocarbon receptor mediate the antiproliferative effects of indirubins. Oncogene, 23: 4400-4412. Koo, J. and Arain, S. (1998). Traditional Chinese medicine for the treatment of dermatologic disorders. Archieves of Dermatology, 134: 1388-1393. Koo, J. and Desai, R. (2003). Traditional Chinese medicine in dermatology. Dermatologic Therapy, 16: 98-105. Lebwohl, M. (2003). Psoriasis. Lancet, 361: 1197-1204. Lin, X.R. (1993). Psoriasis in China. The Journal of Dermatology, 20: 746-755. Lin, Y.K., Yen, H.R., Wong, W.R., Yang, X.H. and Pang, J.H. (2006). Successful treatment of pediatric psoriasis with indigo naturalis composite ointment. Pediatric Dermatology, 23: 507-510. Lin, Y.K, Wong, W.R. and Pang, J.H. (2007). Successful treatment of recalcitrant psoriasis with indigo naturalis ointment. Clinical and Experimental Dermatology, 32: 99-100. Lin, Y.K, Wong, W.R., Chang, Y.C., Chang, C.J., Tsay, P.K, Chang, S.C. and Pang, J.H. (2007). The efficacy and safety of topically applied indigo naturalis ointment in patients with plaque-type psoriasis. Dermatology, 214: 155-16I. Lin, Y.K, Chang, C.J., Chang, Y.C., Wong, W.R., Chang, S.C. and Pang, J.H. (2008). Clinical assessment of patients with recalcitrant psoriasis in a randimized, observer-blind, vehiclecontrolled trial using indigo naturalis. Archives of Dermatology, 144: 1457-1464. Lin, Y.K., Leu, Y.L., Yang, S.H., Chen, H.W., Wang, C.T. and Pang, J.H. (2009). Antipsoriatic effects of indigo naturlis on the proliferation and differentiation of Kerotinozytes with indirubin as the active component. Journal of Dermatoloued Science, 54: 168-174. Marko, D., Schatzle, S., Friedel, A., Genzlinger, A., Zankle, H., Meijer, L. and Eisenbrand, G. (2001). Inhibition of cyclin-dependent kinasel (CDKl) by indirubin derivatives in human tumor cells. Brit~sh Journal of Cancer, 84: 283-289. Nam, S., Buettner, R., Turkson, J., Kim, D., Cheng, J.Q., Muehlbeyer, S., Hippe, F., Vatter, S., Merz, KH., Eisenbrand, G. and Jove, R. (2005). Indirubin derivatives inhibit State signaling and induce apoptosis in human cancer cells. Proceedings of the National Academy of Sciences to the United States of Aermica, 102: 5998-6003. Ortonne, J.P. (1996). Aetiology and pathogenesis of psoriasis. The British Journal of Dermatology, 135: 1-5. Sreepriya, M., Devaki, T. and Nayeem, M. (2001). Protective effects of Indigofera tinctoria L. against D-Galactosamine and carbon tetrachloride challenge on 'in situ' perfused rat liver. Indian Journal of Physiology and Pharmacology, 45: 428-434.
Indigo Naturalis as an Alternative Therapy in Psoriasis vulgaris
43
Suzuki, K, Adachi, R., Hirayama, A., Watanabe, H., Otani, S., Watanabe, Y. and Kasahara, T. (2005). Indirubin, a Chinese anti-leukaemia drug, promotes neutrophilic differentiation of human myelocytic leukaemia HL-60 cells. British Journal Haematology, 130: 681-690. Traub, M. and Marshall, K (2007). Psoriasis-pathophysiology, conventional, and alternative approaches to treatment. Alternative Medicine ReUlew, 12: 319-330. Tse, T.W. (2003). Use of common Chinese herbs in the treatment of psoriasis. Clinical and Experimental Dermatology, 28: 469-75. Verucchi, G., Calza, L., Attard, L. and Chiodo, F. (2002). Acute hepatitis induced by traditional Chinese herbs used in the treatment of psoriasis. Journal of Gastroenterology and Hepatology, 17: 1342-1343. Xiao, Z., Hao, Y., Liu, B. and Qian, L. (2002). Indirubin and meisoindigo in the treatment of chronic myelogenous leukemia in China. Leukemia & Lymphoma, 43: 1763-17688. Yip, S.Y. (1984). The prevalence of psoriasis in the Mongoloid race. Journal of the American Academy of Dermatology, 10: 965-968. Yuan, Z.Z., Yuan, X. and Xu, Z.X. (1982). Studies on tabellae indigo naturalis in treatment of psoriasis. Journal of Traditional Chinese Medicine, 2: 306.
"This page is Intentionally Left Blank"
3 Development of Anti-Angiogenesis Therapies for Treating Cancer from Chinese Medicinal Herbs STEPHEN
M. SAGAR l ,*, RAlMOND K. WONG l
AND DONALD R.YANCE JR.2
ABSTRACT A new pharmacological approach for managing cancer should simultaneously target the multiple biochemical and physiologic pathways that promote cancer progression. Complex mixtures ofphytochemicals used at relatively low doses can reduce tumor adaptation and resistance to conventional cytotoxic chemotherapy, as well as the prevention ofprogression of microscopic disease following definitive therapy. Angiogenesis is a key process in the promotion of cancer. Many plant-derived chemical mixtures manifest both antiangiogenic as well as other anticancer activities. The present article focuses on products that have a high degree of antiangiogenic activity, but it also briefly describes some of the many other actions of these agents that can inhibit tumor progression and reduce the risk of metastases. Natural health products target molecular pathways other than angiogenesis, including epidermal growth factor receptor, the HER2/neu gene, the cyclooxygenase- 2 enzyme, the nuclear factor kappa-B transcription factor, the protein kinases, the Bcl-2 protein, and coagulation pathways. Antiangiogenic Chinese medicinal herbs include Artemisia annua (Chinese wormwood), Curcuma longa (curcumin), Scutellaria baicalensis (Chinese skullcap), Magnolia officinalis (Chinese magnolia tree), Camellia sinensis (green tea), Ginkgo biloba, Poria cocos, Zingiber officinalis (ginger), Panax ginseng, and Rabdosia rubescens hora (Rabdosia). Quality assurance of appropriate extracts is essential prior to embarking upon clinical trials. More data are required on dose-response, appropriate combinations, and 1. Associate Professor, Departments of Oncology and Medicine, McMaster University,
Canada. 2. Center for Natural Healing, Ashland, Oregon. * Corresponding author: E-mail: stephen.saga~cc.hhsc.ca.
46
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
potential toxicities. Given the multiple effects of these agents, their future use for cancer therapy probably lies in synergistic combinations. During active cancer therapy, they should generally be evaluated in combination with chemotherapy and radiation. In this role, they act as modifiers of biologic response or as adaptogens, potentially enhancing the efficacy of the conventional therapies. Key words : Angiogenesis, anti angiogenic, natural health products, herbal medicine, anticancer, molecular biology
INTRODUCTION The Importance of Antiangiogenesis Therapies for Preventing Cancer Progression The induction and promotion of cancer is a multistep process that involves biochemical interactions from the level of the genes, through cell-signaling pathways, intercellular communication mechanisms, supply of nutrients, channels for metastases, and a host immune response. Part of this process is the induction of new blood vessels to supply nutrients and immunocytes. Many natural health products that inhibit angiogenesis also manifest other anticancer activities. This multi-targeted approach may have major advantages for the treatment of cancer by overcoming the therapeutic resistance to the single drug approach caused by multiple redundant physiological pathways in the cancer cell. Cancer progression requires a source of nutrition and oxygen. Tumors that outgrow their oxygen supply cannot form masses more than 1 to 2 mm in size or they then develop central necrosis. Neoplasms are genetically plastic and often adapt by switching on genes that result in an increased ability to invade and to metastasize. A critical part of this process is the induction of local small blood vessels, termed angiogenesis (Fidler, 2000a; Fidler, 2000b). Tumors do not grow progressively unless they induce a blood supply from the surrounding stroma. Cancers that lack angiogenesis remain dormant. Rapid logarithmic growth follows the acquisition of a blood supply. The tumor angiogenic switch is activated (Fig 1) when the balance of angiogenic inhibitors to stimulators is shifted (Wallace, 2002). The process of neovascularization is subtly controlled in normal tissues by a series of endogenous polypeptides that are secreted during growth, healing, and tissue renewal (Table 1). Cancers synthesize or induce some of these polypeptides, especially vascular endothelial growth factor (VEGF) and angiopoietin (APN). These are peptides that are stimulated by hypoxia and result in sprouting of endothelial cords. The cancer induces a profuse but immature network of thin endothelial-lined channels, essential for tumor oxygenation. Although these new vessels allow progressive tumor growth, they are less efficient than the vascular supply of normal tissues.
Anti-Angiogenesis Therapies for Treating Cancer
47
APN normally recruits pericytes and initiates modeling of the vessel wall to more mature forms. However, tumors secrete a relative excess ofVEGF, and this results in disorganized and leaky vessels that cause local bleeding and edema. Antiangiogenic therapy is less susceptible to the development of treatment resistance because it is directed to stromal tissue rather than the genomically unstable tumor cells. Table 1. Endogenous angiogenic polypeptides • • • • • • • • • • • • • • • • • • •
Angiogenin (AG) and Angiotropin (AT) Basic Fibroblast Growth Factor (bFGF) Granulocyte-Colony-Stimulating Factor (G-CSF) Hepatocyte Growth Factor (HGF) Interleukin-8 (11-8) Placental Growth Factor (PGF) Platelet-Derived Endothelial Cell Growth Factor (PD-ECGF) Pleiotrophin (PTN) Proliferin Transforming Growth Factor-a (TGF-a) Transforming Growth Factor-13 (TGF-13) Tumour Necrosis Factor-a (TNF-a) Vascular Endothelial Growth Factor (VEGF) Vascular Permeability Factor (VPF) Insulin-Like Growth Factor I and II (IGF-l and II) Cyclooxygenase (COX) and Lipoxygenase (LOX) Nuclear Factor-Kappa 13 (NF-KB) Activator Protein (AP-l) Angiopoietin (APN)
• Tumour angiogenic switch is activated when balance of angiogenic inhibitors to stimulators is shifted • Reduced production of thrombospondin (TSP-l) from fibroblasts • Vascular endothelial factor (VEGF) stimulates new vessel formation but exceeds Angiopoietin (APN) that normally induces differentiation (pericytes) • Matrix metalloproteinase induction • Increased VEGF relative to APN results in disorganized leaky vessels • Rapid logarithmic growth follows the acquisition of a blood supply
Fig 1. The angiogenic switch
Single antiangiogenic agents, such as monoclonal antibodies or specific tyrosine kinase inhibitors, seem to have limited efficacy. Natural health products contain a range of complex organic chemicals that have synergistic activity. They inhibit angiogenesis by interacting with multiple pathways, as well as having other activities that can interact with cell signaling, the apoptotic pathway, and the interaction of cancer cells with the immune system. Some antiangiogenic agents also have anticoagulation activity that may also be associated with a reduction of metastases. Heparin is a well known example of a therapy with both anticoagulation and antiangiogenic activities. Instead of developing multiple monoclonal antibodies to target
48
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
the various peptides and their receptors, an alternative approach would be to evaluate phytochemicals that influence multiple pathways. The science of pharmacognosy evaluates drugs derived from herbal remedies or phytomedicines. There has been minimal clinical research that evaluates their use as adjuvant therapy to conventional treatment with cytotoxic drugs and radiotherapy. Antiangiogenic natural health products may be most effective in impeding cancer recurrence after cytotoxic therapy, encouraging tumors to remain dormant by changing the balance from cell proliferation to cell death by apoptosis.
The Angiogenic-Metastatic Pathway as a Target for Anticancer Therapies The process of cancer metastasis consists of a series of sequential interrelated steps. Each step is rate limited and may be a target for therapy. The outcome of the process is dependent on both the intrinsic properties of the tumor cells and the responses of the host. The balance of these interactions varies between tumors and patients. The major steps in the formation of a metastasis are as follows (Sugarbaker, 1979; Hart et al., 1989; Liotta & Stetler-Stevenson, 1991): 1. Transformation of normal cells into tumor cells followed by growth. Initially depends on nutrients supplied by simple diffusion. 2. Extensive vascularization (angiogenesis). This must occur if the tumor mass is to exceed 1 mm in diameter. The production and secretion of proangiogenic factors by tumor cells and host cells play a major role in establishing a capillary network from the surrounding host tissue. 3. Local invasion. Tumor cells invade the host stroma through several mechanisms. Thin-walled venules, fragmented arterioles, and lymphatic channels offer little resistance to penetration and provide the most common pathways for tumor cell entry into the circulation. 4. Detachment and embolization. Single cells or clumps break away. Most circulating tumor cells are rapidly destroyed. Those that survive must arrest in the capillary beds of distant organs by adhering either to capillary endothelial cells or to the exposed subendothelial basement membrane. 5. Extravasation into new host organ or tissue. 6. Proliferation within the new host organ or tissue. To continue growing beyond the size of 1 mm in diameter, the micrometastasis must develop a vascular network and evade destruction by host defenses. The cells can then continue to invade blood vessels, enter the circulation, and produce additional metastases. The growth of many cancers is associated with the absence of the endogenous inhibitors of angiogenesis, such as interferon-beta (INF -~). INF-
Anti-Angiogenesis Therapies for Treating Cancer
49
~ is a potent inhibitor of angiogenesis through blocking interleukin-8 (IL8), basic fibroblast growth factor (bFGF), and collagenase type V, which are all potent angiogenic factors that aid tumor development and invasiveness. VEGF stimulates the proliferation and migration of endothelial cells and induces the expression ofmetalloproteinases and plasminogen activity. Over expression of VEGF in tumor cells enhances tumor growth and metastasis in several animal models by stimulating vascularization (Folkman & Kotran, 1976; Senger et al., 1983; Folkman et al., 1986; Folkman & Klagsbrun, 1987; Nagy et al., 1989; Folkman et al., 1995; Ferrera et al., 1996; Risau, 1997; Kumar et al., 1998). Some cytotoxic chemotherapy agents are being used at lower than normal doses with the intent of inhibiting angiogenesis and minimizing toxicity (Oliff et al., 1996; Miller et al., 2001). This strategy may permit advanced cancer patients to maintain a better quality of life. The low-dose therapy is termed metronomic dosing (Hanahan et al., 2000; Maraveyas et al., 2005; Hudis, 2005). The metronomic model of conventional cytotoxic chemotherapy suggests that there may also be advantages for administering combinations of phytochemicals that interact with the multistep process of angiogenesis (Singh et al., 2003). In other words, targeting the vascular endothelium with continuous low-dose non-cytotoxic therapies may maintain tumor control without excessive toxicity. Their potential role for increasing overall survival (but not necessarily diseasefree survival) and maintaining quality of life requires evaluation in future clinical trials.
Screening Herbs for Antiangiogenic Activity One ofthe first isolated antiangiogenic agents was a phytochemical. Ingber et al. (1990) reported the antiangiogenic properties of fumagillin, a secreted antibiotic of the fungus Aspergillus fumigatus (Trichocomaceae). Refined fumagillin produces excess toxicity, so analogues of fumagillin were subsequently synthesized. Fumagillin and an analogue labeled TNP-470 are proposed to inhibit angiogenesis by selective inhibition of methionine aminopeptidase type 2 (MetAP-2). However, TNP-470 also demonstrated poor pharmacokinetic behavior and dose-limiting toxicity in clinical trials, and these factors remain obstacles to its use as an anticancer agent. Further modifications of fumagillin have been conducted to develop MetAP-2 inhibitors with desirable pharmacological properties. They have been tested only by in vitro assays, and to date, no clinical trials of these analogues have yet been conducted (Ingber et al., 1990; Pyun et al., 2004; Furness et al., 2005). Since the angiogenic cascade is a multistep process, numerous assays have been developed to study potential angiogenic activity. Some analyze a single step in the pathway, whereas others test the angiogenic cascade as a whole. The relationship of each assay to clinical activity is poorly defined. Some agents have profound anti angiogenic effects at low doses; others exhibit anti angiogenic activity only at near cytotoxic concentrations. Some agents have activity in one model but none in others.
50
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Criteria for anti angiogenic activity are listed in Fig 4 and should include (Miller et al., 2001): • differential cytotoxicity, • alteration of endothelial cell function, • critical mechanistic effects, and • inhibition of angiogenesis in vivo. Western Biomedicine
Traditional Chinese Medicine • Stagnation of blood and Qi
• Destagnation herbs
• Increased fluid content and abnormal vascular supply; blood stasis; poor oxygenation • Anticoagulants as adjunct to chemotherapy increase survival in clinical studies; prevent blood-borne metastases in animal models
Fig 2. The traditional chinese medicine versus the biomedical theory of cancer
Various assays (Table 2) are used to screen natural health products for antiangiogenic activity (Miller et al., 2001; Wang et al., 2004; Kruger et al., 2001). Assays used for screening will be briefly discussed. Table 2. Screening for anti-angiogenesis
• In vitro Assays - Human umbilical vein endothelial cells (HUVEC) - Bovine aortic endothelial cells (BAEC) - Tumour endothelial cells (TEC) • In vivo Assays - Chick embryo chorioallantoic membrane (CAM) model - Cornea implantation - Matrigel - Tumour xenografts
In vitro Assays In vitro assays are designed to recapitulate each of the multiple events that constitute the angiogenic process. Some of them are very specific in analyzing a single event (proliferation, apoptosis, migration, production of proteases), whereas others provide a more complex picture of the process involving cell functions and interactions with the environment. In vitro assays for the activity of anti angiogenic compounds are usually based on the use of endothelial cells. A critical issue in setting up an in vitro assay is the choice of endothelial cells. Immortalized endothelial cells are sometimes used, as they provide an "unlimited" source of cells. Although these cell lines have the obvious advantages of being easy to grow and relatively stable
Anti-Angiogenesis Therapies for Treating Cancer
51
throughout in vitro passages and among batches, they have usually lost some of the characteristics of endothelial cells, including molecular markers, and exhibit changes in function. The most commonly used endothelial cells are from the human umbilical vein, since these are easily available and cell isolation is relatively simple. For the same reasons, bovine or murine aortic endothelial cells are often used too, but these come from large vessels, and they have different phenotypic and behavioral characteristics from those of the microvessels that are more likely involved in angiogenesis. Other common sources of microvascular endothelial cells are the skin, brain, adipose tissue, and adrenal gland. Endothelial cells derived from the microvasculature of different tissues/organs are often heterogeneous, imposing a further constraint on the choice of cell model. Ideally, when developing inhibitors of tumor angiogenesis, tumor-derived endothelial cells should be used. However, practical difficulties in their isolation from tumor tissue and maintenance in culture have limited their use in preclinical studies (Taraboletti et al., 2004). The ability to maintain endothelial cells in culture has allowed the study of endothelial cell proliferation, migration, and cellular function. Angiogenic activity may be represented as endothelial cell migration across a Boyden chamber. Compounds with antiangiogenic potential will inhibit the migration. The bovine aortic endothelial cell (BAEC) and the human umbilical vein endothelial cell (HUVEC) assays are established systems. In vitro assays are relatively inexpensive and give more rapid results. However, the ability to inhibit endothelial cell proliferation, migration, and tubule formation in vitro may not necessarily predict in vivo response. In vitro assays are a rapid method for initial screening of large numbers of agents. Definitive conclusions cannot be based on in vitro assays alone.
In vivo Assays These biological assays are more specific for detecting antiangiogenic activity. The chick embryo chorioallantoic membrane (CAM) model is an extraembryonic membrane that is commonly used to study agents that influence angiogenesis. An angiogenic response occurs 72 to 96 h after stimulation in the form of increased vessel density around the implant. On the other hand, an angiostatic compound induces the vessels to become less dense around the implant and even disappear. Other systems include animal cornea implantation, disc angiogenesis, Matrigel systems, and tumor xenograft models. The in vivo assays provide a more complete physiologic assessment of angiogenesis but are more time-consuming and expensive.
Criteria for Antiangiogenic Activity The degree of anti angiogenic activity is dose dependent. Most chemotherapy drugs have antiangiogenic activity when administered at high doses. We
52
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
are especially interested in compounds that specifically interact and antagonize the steps involved in angiogenesis when administered at low doses. These agents may have relatively low toxicity at low dose and may exhibit a higher therapeutic gain. Most conventional chemotherapy drugs have some degree of anti angiogenic activity as a consequence of their cytotoxic activity. Ideal botanical derivatives would specifically antagonize new vessel formation in tumors, without significant toxicity to normal tissues and without major adverse reactions. The ideal agent would also inhibit tumor cell proliferation through other physiologic pathways, such as influencing intracellular signaling pathways. Multiple levels of antiangiogenic activity may be required to overcome the development of resistance by tumor-associated endothelial cells (TEC). Survival factors, such as the increased secretion of VEGF and bFGF by the tumor cells, activate intracellular pathways that prevent TEC apoptosis. Maximal anti angiogenic activity usually requires prolonged exposure to low concentrations ofthe active agent. This approach contrasts with the concept of administering maximum tolerated doses of cytotoxic drugs to maximize tumor cell kill. Some reports have confirmed the utility of combining low, frequent-dose chemotherapy plus an agent that specifically targets the endothelial cell compartment (Hanahan et al., 2000; Maraveyas et al., 2005). The evidence suggests that an anti angiogenic schedule can be more effective than using high-dose cytotoxic drugs alone. We hypothesize that concomitant scheduling of antiangiogenic botanicals with low, frequentdose cytotoxic therapies may have biological advantages that can increase therapeutic gain.
Natural Health Products that Inhibit Angiogenesis Further research is necessary to screen herbs that may be useful anti angiogenic therapies. Table 3 lists Chinese herbs with anti angiogenic activity (Wang et al., 2004), and Table 4 lists herbs and their derivatives that inhibit VEGF (Singh et al., 2003). An expert herbalist can advise on potential herbal treatments derived from advanced traditional medical systems, such as traditional Chinese medicine. It will be imperative to develop a new model of modern pharmacology based on traditional pharmacognosy. Our developing knowledge of cancer biology suggests that administering cytotoxic drug therapy at very high doses is not always appropriate. A new approach is to administer lower doses of synergistic organic chemicals. These complexes already exist in myriad botanicals. New laboratory techniques allow more specific assays of activity and enable quality assurance and consistency between batches of botanical preparations to be maintained. This will enable credible clinical trials of antiangiogenic natural health products to be initiated.
Anti-Angiogenesis Therapies for Treating Cancer
53
Table 3. Antiangiogenesis activity of chinese medicinal herbal extracts (Exhibiting more than 20% inhibition at 0.2 g/herb/mL) (Wang et al., 2004) Assays: chick embryo chorioallantoic membrane (CAM) and bovine aortic endothelial cells culture models (BAEC) Used Part
Name
Berberis paraspecta Catharanthus roseus Coptis chinensis Scrophularia ningpoensis Scutellaria baicalensis Polygonum cuspidatum Taxus chinensis
% Inhibition
% Inhibition
(CAM)
(BAEC)
25 27 25 20 27
38 30 37 34 41 28
Root Leaf Rhizome Root Root Whole plant Bark
26
Traditional Chinese Medical (TCM) theory describes tumor formation as stagnation of qi and blood (Fig 2). In Western theory, this corresponds to the abnormal vasculature induced by the cancer when it induces the angiogenic switch. The tumor has a stagnant blood supply with abnormal leaky vessels that raises the interstitial fluid pressure, resulting in centrifugal diffusion and poor oxygenation. TCM theory suggests "destagnation herbs" as a potential therapy. A randomized placebocontrolled trial from China showed that the addition of "de stagnation" herbs (including Salvia miltiorrhiza Bunge [Lamiaceael and Angelica sinensis Diels [Apiaceae]) to radiotherapy doubled both the local control and survival rates of patients with nasopharyngeal cancer (Fig 3) (Xu et al., 1989). In some clinical trials, anticoagulation drugs are associated with a reduction in metastases (Hejna et al., 1999; Smorenburg et al., 2001; Blom et al., 2005). Laboratory evidence now suggests that many of these Chinese herbs have anti angiogenic and anticoagulation properties (Huang et al., 2003; Wang et al., 2004; Samuels, 2005).
Destagnation herbs (Huo Xue Hua Yu) activate blood flow and improve micro-circulation (huang qi, chi shao, chuan xiong, dang gui, tao ren, hong hua, ji xue teng, ge gen, chen pi, dan shen) N XRT alone XRT + herbs
90 98
Local recurrence
5 Year survival
29% 14% (p<0.05)
37% 53% (p<0.05)
Fig 3. A randomized controlled trial of chinese herb de stagnation formula with radiation in the treatment of nasopharyngeal carcinoma (Xu et al., 1989)
54
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Some herbs that are traditionally used in China as anticancer agents have been screened for their antiangiogenic activity (Fig 4) in the laboratory (Wang et ai., 2004). Table 3 lists the most active herbs (exhibiting more than 20% inhibition at 0.2 g/herb/mL), using the CAM and BAEC assays. Other herbs are included in Table 4 and are discussed in more detail. Table 4. Chinese herbs and their derivatives that specifically inhibit vascular endothelial growth factor and have direct activity against angiogenesis
• Artemisia annua (Chinese wormwood; contains 95% artemisinin and other related terpenes and flavonoids) • Scutellaria baicalensis (Chinese Baical skullcap; contains 95% baicalin and flavonoids) • Curcuma Zonga (turmeric; contains 95% curcumin) • Camellia sinensis (green tea; contains 95% phenols; 50% epigallocatechin) • Angelica sinensis (Dong quai; contains 4-hydroxyderricin) • Taxus brevifolia (Pacific yew; contains taxol) • Polygonum cuspidatum (Japanese knotweed; contains 20% resveratrol) • Silybum marianum (Milk thistle; contains 80% silymarin [silibin]) • Ginkgo biloba • Magnolia obovata (contains 90% honokiol) • Zingiber officinale (contains 6-gingerol) • Poria cocus • Panax ginseng • Rabdosia rubescens (Rabdosia) • Berberis paraspecta Ahrendt (Berberidaceae), root • Catharanthus roseus G Don (Apocynaceae), leaf • Coptis chinensis Franch (Ranunculaceae), rhizome • Scrophularia ningpoensis Hemsl (Scrophulariaceae), root • Polygonum cuspidatum, Whole plant • Taxus chinensis Rehder (Taxaceae) • Degree of antiangiogenic activity is dose dependent • Specifically interacts and antagonizes angiogenesis at low doses (high therapeutic gain) • Inhibits tumor cell proliferation through multiple physiologic routes, such as intracellular signaling pathways • Minimal effect on normal angiogenesis • Low toxicity with prolonged administration Fig 4. Criteria for an anti-angiogenic botanical
Botanicals usually act on multiple anticancer targets since they contain a variety of organic chemical complexes. The biochemical signaling pathways of angiogenesis form a complex, interconnected web. Inhibition of one part of this web may result in compensation through another pathway. A potential advantage of phytochemicals is that they may act through multiple pathways and reduce the development of resistance by cancer cells. This model of pharmacognosy recognizes the advantage of administering the whole plant product to maximize activity (Fig 5). Over extraction of a specific chemical constituent may remove this therapeutic gain. The
Anti-Angiogenesis Therapies for Treating Cancer
55
challenge for modern pharmacognosy is to ensure that the optimum mixture of chemical constituents is maintained when purifying the product. Usually, this will require a combination of both chemical and biological assays. Further anticancer properties of some anti angiogenic botanicals will briefly be discussed. Their effects may also interact with various biochemical pathways that indirectly influence angiogenesis. Traditional practice has been to combine multiple natural health products, and this may scientifically provide a therapeutic advantage. • Range of complex organic chemicals that may have synergistic activity, alone or in combinations. • Inhibit angiogenesis by interacting with multiple pathways. • Bonus activities: interact with cell signaling, apoptosis and interaction of cancer cells with the immune system. • Anticoagulation activity that may be associated with a reduction of metastases • Less acquired resistance to therapy. • Low toxicity: suitable for combination therapy and long term administration. • Possible economic advantages. Fig 5. Advantages of anti-angiogenic NHPs
Chinese Medicinal Botanicals for Potential Development into AntiAngiogenic Therapeutic Agents for the Treatment of Cancer
Artemisia annua (Chinese Wormwood) Artemisinin is the active constituent extracted from the plant A. annua L. (Asteraceae). It has been used clinically as an antimalarial drug (Mueller et al., 2004). More recently, it was shown to be cytotoxic to cancer cells through induction of apoptosis (Singh & Lai, 2004). Artesunate (ART) is a semisynthetic derivative of artemisinin. The in vitro effect of ART was tested on the HUVEC model of angiogenesis. It significantly inhibited angiogenesis in a dose-dependent manner. The inhibition of HUVEC proliferation was greater than the effect on cancer cells, fibroblast cells, and human endometrial cells. This indicates that its anti angiogenic activity is greater than its cytotoxicity. The antiangiogenic effect in vivo was evaluated in nude mice using transplanted human ovarian cancer (HO891) cells and immunohistochemical staining for microvessel CD31 antigen, VEGF, and the VEGF receptor (KDRlflk-1). Tumor growth was decreased and microvessel density was reduced without any toxicity to the host animals. Artemisinin also lowered the VEGF expression by tumor cells and the KDRlflk-1 expression by endothelial cells (Chen & Chang, 2004). Artemisinin also has therapeutic effects through other pathways. It inhibits the activation of nuclear factor-KB (NF-KB), an important activator protein in cancer development and progression (Aldieri et al., 2003).
Scutellaria baicalensis (Chinese Skullcap) Baicalin and baicalein are the main derivatives from the Chinese Skullcap herb, S. baicalensis Georgi (Lamiaceae). They are potent anti angiogenic
56
Compo Bio. Nut. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation!
compounds that reduce VEGF, bFGF, 12-lipoxygenase activity, and MMP (Liu et aZ., 2003; Miocinovic et aZ., 2005). It is a component of the traditional Bupleurum formula for stagnation (Xiao Chai Hu Tang): • Bupleurum (Chai Hu) 12-15 g BupZeurum chinense • Scutellaria (Huang Qin) 9-12 g Scutellaria baicaZensis • Pinelli a (Ban Xia) 9-12 g Pinellia ternata • Fresh Ginger (Sheng Jiang) 3-6 g Zingiberis offici naZis • Ginseng (Ren Shen) 6-9 g Panax ginseng • Licorice (Gan Cao) 3-6 g GZycyrrhiza uraZensis • Jujube Dates (Da Zao) 3-5 pcs Zizyphus jujuba S. baicaZensis is also one of the herbs found in PC-SPES, a complex of Chinese herbs that may have clinical activity against advanced prostate cancer (Small et aZ., 2000; Oh et aZ., 2001). However, although a phase 2 trial demonstrated clinical activity in patients with androgen independent prostate cancer, diethylstilbestrol (DES) and ethinyl estradiol (both known to have potent antiprostate cancer activity) were detected in various lots of PC-SPES (Hsieh et aZ., 2002). It is still intriguing that the decline in PSA was greater for PC-SPES that was potentially contaminated with DES than for the comparator group that received DES alone, suggesting some independent activity. Although baicalin and baicalein have multiple anticancer activities in vitro, their clinical activity is not established, and their contribution to any potential therapeutic effect of PC-SPES is unknown (Cordell, 2002; Oh et aZ., 2004).
Curcuma longa (Turmeric) Curcumin is the most active curcuminoid present in turmeric, C. Zonga L. (Zingiberaceae). It interacts with cancer cells at a number of levels and can enhance the tumoricidal efficacy of cytotoxic chemotherapy and radiotherapy (Narayan, 2004; Khafif et aZ., 2005; Sen et aZ., 2005). Its antiinvasive effects are partly mediated through the down regulation of MMP2 and the upregulation of tissue inhibitor of metalloproteinase (TIMP-l) (Shao et aZ., 2002). These enzymes are involved in the regulation of tumor cell invasion. Curcumin inhibits the transcription of 2 major angiogenesis factors, VEGF and bFGF (Arbiser et aZ., 1998). It interacts with VEGF and nitric oxide- mediated angiogenesis in tumors (Sreejayan, 1997; GarciaCardena et aZ., 1998). Elevated levels of nitric oxide correlate with tumor growth. Curcumin reduces nitric oxide generation in endothelial cells. CD13/ aminopeptidase-N (APN) is a membrane bound enzyme found in blood vessels undergoing active angiogenesis. Curcumin binds to APN and blocks its activity, thereby inhibiting angiogenesis and tumor cell invasion (Gururaj et aZ., 2002; Shim et aZ., 2003). Derivatives of curcumin may be
Anti-Angiogenesis Therapies for Treating Cancer
57
developed to target APN, providing a novel approach to reduce neoplastic angiogenesis (John et al., 2002; Hahm et al., 2004). Curcumin also downregulates the expression of the VEGF and MMP-9 genes that are associated with angiogenesis. Demethoxycurcumin is a structural analogue of curcumin isolated from Curcuma aromatica Salisb. (Zingiberaceae). It specifically inhibits the expression of MMP-9 (Kim et al., 2002). Curcumin can interfere with the activity of both MMP-2 and MMP-9, the basis of the angiogenic switch, thereby reducing the degradation of the extracellular matrix (Chen et al., 2004). It also interferes with the release of angiogenic factors that are stored in the extracellular matrix. It inhibits growth factor receptors such as epidermal growth factor receptor (EGFR) and VEGFR and the intracellular signaling tyrosine kinases. This cell-signaling system can promote further angiogenesis through gene activation that increases levels of cyclooxygenase-2 (COX-2), VEGF, IL-8, and the MMPs (Reddy & Aggarwal, 1994; Dorai et al., 2001; Leu et al., 2003). A phase 1 study of curcumin found no treatment-related toxicity up to 8000 mg/d. Beyond 8000 mg/d, the bulky volume of the drug was unacceptable to the patients. The serum concentration of curcumin usually peaked at 1 to 2 h after oral intake of curcumin and gradually declined within 12 h (Cheng et al., 2001). This study suggested that it may prevent cancer progression. Derivatives of curcumin, such as copper chelates of curcuminoids, may have increased antitumor activity (John et al., 2002).
Camellia sinensis (Green Tea) C. sinensis (L.) Kuntze (Theaceae), contains polyphenols and catechins (mainly epigallocatechin-3 gallate [EGCG]) (Lee et al., 2002). These constituents inhibit MDAMB breast cancer cell and HUVEC proliferation (Sartippour et al., 2002). In addition, they suppress breast cancer xenograft growth and reduce the density of tumor vessels in rodent studies (Cao & Cao, 1999). This is associated with a decrease in VEGF, regulated at the level of transcription. EGCG also suppresses protein kinase C (PKC), another VEGF transcription modulator. Inhibition of VEGF transcription is one of the molecular mechanisms involved in the anti angiogenic effects of green tea that may contribute to its potential use for cancer treatment (Kojima-Yuasa et al., 2003; Tang et al., 2003; Fassina et al., 2004). EGCG may be administered as a powdered extract of green tea. An appropriate dose has been extrapolated from antiangiogenic activity in rodent experiments as well as a phase 1 study in humans (Cao et al., 1999; Pisters et al., 2001). A dose of 1.0 g/m3 times daily (equivalent to 7-8 Japanese cups [120 mLl 3 times daily) has been recommended. In practice, lower total daily doses of 2 to 4 g of standardized green tea extract (95% polyphenols/60% catechins) are usually prescribed. Each gram of this extract provides 400 to 500 mg of EGCG. The dose-limiting adverse effects are gastrointestinal and
58
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
neurological effects of caffeine. However, the caffeine may potentiate the antiangiogenic effect of EGCG (Pisters et al., 2001).
Polygonum cuspidatum (Japanese Knotweed) This contains 20% resveratrol. It has anti angiogenic activity demonstrated by its ability to inhibit HUVEC division and to decrease the lytic activity of MMP-2 (Cao et al., 2005). Resveratrol inhibits VEGF induced angiogenesis by disruption of reactive oxygen species-dependent Src kinase activation and subsequent VE-cadherin tyrosine phosphorylation (Igura et al., 2001; Lin et al., 2003). Resveratrol inhibits the growth of gliomas in rats by suppressing angiogenesis (Tseng et al., 2004).
Silybum marianum (Milk Thistle) Silibinin and silymarin are polyphenolic flavonoids isolated from the fruits or seeds of S. marianum (L.) Gaertn. (Asteraceae). In the laboratory, silymarin demonstrates strong activity against a variety of tumors through down regulating VEGF and EGFR (Jiang et al., 2002; Singh & Aggarwal, 2003). Silymarin suppresses VEGF when used as a single agent against human ovarian cancer endothelial cells in vitro (Gallo et al., 2003). A more detailed discussion can be found elsewhere (Sagar, 2007).
Ginkgo biloba G. biloba L. (Ginkgoaceae) extract has anticancer effects that are related to its gene-regulatory and antiangiogenic properties. The G. biloba extract used in most of the research is EGb 761, which contains about 25% flavonoids (ginkgo-flavone glycosides) and about 5% terpenoids (ginkgolides and bilobalides). The most potent flavonoid is ginkgolide B. This extract inhibits angiogenesis by down regulating VEGF (Zhang et al., 2002; De Feudis et al., 2003).
Magnolia officinalis (Chinese Magnolia Tree) The seed cones of M. officinalis Rehder & E.H. Wilson (Magnoliaceae) contain substances that inhibit the growth of new blood vessels. Honokiol is the active constituent. It may partly reduce angiogenesis through the regulation of platelet-derived endothelial cell growth factor and TGF-f3 expression. It also inhibits nitric oxide synthesis and TNF-f3 expression (Son et al., 2000; Lee et al., 2004). In animal experiments, it suppresses the proliferation of blood vessel endothelial cells more than other types of cells and thereby reduces tumor growth (Bai et al., 2000; Chen et al., 2004).
Zingiber officinale (Ginger) 6-Gingerol, fromZ. officinale Roscoe (Zingiberaceae), inhibits both the VEGFand bFGF-induced proliferation of human endothelial cells and causes cell cycle arrest. It also blocks capillary-like tube formation by endothelial cells in response to VEGF and strongly inhibits sprouting of endothelial cells in
Anti·Ang iogenesis Therapies for Treating Cancer
59
the rat aorta and mouse cornea in vitro models. In mice receiving injections of B16F10 melanoma cells, intraperitoneal administration of 6-gingerol, at doses less than cytotoxic levels, reduces the number of lung metastases (Kim et al., 2005).
Poria cocos P. cocos F.A. Wolff (Coriolaceae) is a mushroom extract that has been traditionally held to have anticancer activity. It inhibits platelet aggregation
and appears to be antiangiogenic by downregulating NF-kappa-B (Jin et al., 2003; Chen et al., 2004; Lee et al., 2004).
Panax ginseng The lipophilic constituents of P. ginseng C.A. Meyer (Araliaceae) are called saponins (or ginsenosides). These extracts possess anticancer activities in tumors that include antiangiogenesis and induction of tumor cell apoptosis (Sato et al., 1994).
Rabdosia rubescens (Rabdosia) The herb R. rubescens H . Hara (Lamiaceae) is used traditionally to treat cancer and is a constituent of the PC-SPES formula. It contains ponicidin and oridonin, two diterpenoids that possess significant antiangiogenic activity (Meade-Tollin et al., 2004).
CONCLUSIONS Angiogenesis involves multiple interdependent processes operating at the molecular level. These include gene expression, signal processing, and enzyme activities. Most antiangiogenic natural health products block new vessel formation at multiple levels. Lack of standardization of screening assays may be an obstacle to defining the most effective products for clinical use. Over extraction of constituents may negate some of the advantages of potential synergy. Mainly preclinical data exist for most of the naturally derived anti angiogenic agents. Most of the studies of anti angiogenic activity are based on in vitro or animal work, which cannot be readily extrapolated to humans. Phase 1 and 2 studies are required to determine their potential to improve cytotoxic therapies. Quality assurance of appropriate extracts is essential prior to embarking on clinical trials (Fig 6). Since anti angiogenic agents are mainly cytostatic in nature, the usual paradigm for anticancer drug development, in which tumor response in phase 2 trials prompts further development, is not always appropriate. More data are required on dose response, appropriate combinations, and potential toxicities. Given the multiple effects of these agents, their future use for cancer therapy probably lies in synergistic combinations. They may
60
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
4·
wi
Traditional Qbs~tvati!:>nS'
Authentication ;lmf;f~oslational research
Dose optimization
Phase I
effi l'ldverse taifects
Pha.se ,Ill ral)dQ.tniZ~ placebo oontrolled "" ;!" efficacy studY '0
Fig 6. Systematic research of natural health products
be evaluated alone for the prevention of cancer recurrence following definitive treatment. To be suitable for long-term chronic use, these agents should possess minimal toxicity and should be orally administered. However, angiogenesis is also essential for healing of injuries. Most compounds that inhibit tumor angiogenesis are likely to inhibit physiologic angiogenesis, leading to potential side effects, such as ulceration and bleeding. Studies are required to determine distinguishing features of tumor vessels from normal vessels to enable a therapeutic gain to be achieved. During active cancer therapy, they should generally be evaluated in combination with chemotherapy and radiation. In this role, they act as biological response modifiers and adaptogens, potentially enhancing the efficacy of the so-called conventional therapies. The diversity of angiogenic factor expression in different tumors receiving various therapies, combined with the fact that endothelial cells in different tumors are phenotypically distinct, is a major challenge for the development of effective antiangiogenic regimens (Jung et at., 2000; Sweeney et at., 2003). Their effectiveness may be increased when multiple agents are used in optimal combinations. Surrogate markers, such as angiogenic cytokines, are necessary to predict anti angiogenic response. Circulating levels of FGF-2, VEGF, vascular adhesion molecule (V-CAM-1), endothelial intercellular adhesion molecule (ICAM-l), IGF-1, and cytokines such as IL-8 may correlate with tumor angiogenesis (Yoshida et at., 1997; Tang et at., 2001; Brower, 2003; Ruegg, et at., 2003; Ria et ai., 2004). In addition, circulating endothelial cells and their progenitors may be a more reliable marker of response to anti angiogenic therapies (Salcedo et at., 2005; Schneider et ai., 2005; Shaked et ai., 2005). Noninvasive functional imaging, such as positron emission tomography and functional magnetic resonance imaging, may playa role (Neeman, 2000).
Anti-Angiogenesis Therapies for Treating Cancer
61
Current laboratory evidence suggests a useful role for natural health products in the treatment of cancer. The input of an ethnobotanist, oncologist, laboratory scientist, and a clinical trials methodologist to the research effort is essential to distill the wealth of traditional knowledge into a modern framework that can be evaluated scientifically. The determination of the biologically active dose that may possess least toxicity may be relevant. Combinations of whole herbs or constituent phytochemicals at lower doses are important. In addition, a longer period of exposure to the natural health product may be more effective than a short exposure to the highest possible dose level. New designs for trials to demonstrate activity in human subjects are required. Although controlled trials might be preferred, smaller studies with appropriate end points and surrogate markers for anti angiogenic response could help prioritize agents for the larger resource-intensive phase 3 trials. Because most of the agents are expected to be cytostatic, it is inappropriate to require the standard criteria of measured tumor response. On the other hand, simply confirming stable disease may be misleading. More research on surrogate markers of antiangiogenic response is obviously necessary prior to directing resources to large-scale clinical trials. A multidisciplinary approach by the herbalist and the oncologist is important for implementing and studying natural health products used for the treatment of patients with cancer. We now have a better understanding of their effects at the molecular level. Introducing these interventions into the clinic through appropriate studies will provide more definitive evidence of efficacy and hopefully improve outcome for many cancer patients.
REFERENCES Aldieri, E., Atragene, D. and Bergandi, L. (2003). Artemisinin inhibits inducible nitric oxide synthase and nuclear factor NF-KB activation FEBS Lett., 552: 141-144. Arbiser, J.L., Klauber, N. and Rohan, R. (1998). Curcumin is an in vivo inhibitor of angiogenesis Mol Med., 4: 376-383. Bai, X., Cerimele, F. and Ushio-Fukai, M. (2000) Honokiol, a small molecular weight natural product, inhibits angiogenesis in mtro and tumour growth in vivo. J BioI Chem., 278: 35501-35507. Blom, J.W., Doggen, C.J., Osanto, S. and Rosendaal, F.R. (2005). Malignancies, prothrombotic mutations, and the risk of venous thrombosis. JAMA, 293: 715-722. Brower, V. (2003). Evidence of efficacy: researchers investigating markers for angiogenesis inhibitors. J Natl Cancer [nst., 95: 1425-1427. Cao, Y. and Cao, R. (1999). Angiogenesis inhibited by drinking tea. Nature, 398: 381. Cao, Y., Fu, Z.D., Wang, F., Liu, H.Y. and Han, R. (2005). Anti-angiogenic activity of resveratrol, a natural compound from medicinal plants. J Asian Nat Prod Res., 7: 205-213. Chen, F., Wang, T. and Wu, Y.F. (2004). Honokiol: a potent chemotherapy candidate for human colorectal carcinoma. World J Gastroenterol., 10: 3459-3463. Chen, H.H., Zhou, H.J., Wu, G.D. and Lou, X.E. (2004). Inhibitory effects of artesunate on angiogenesis and on expressions of vascular endothelial growth factor and VEGFreceptor KDRlflk-1. Pharmacology, 7: 1-9. Chen, H.W., Yu, S.L. and Chen, J.J. (2004). Anti-invasive gene expression profile of curcumin in lung adenocarcinoma based on a high throughput microarray analysis. Mol Pharmacol., 65: 99-110.
62
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation!
Chen, Y.Y. and Chang, H.M. (2004). Antiproliferative and differentiating effects of polysaccharide fraction from fu-ling (Poria cocos) on human leukemic U937 and HL60 cells. Food, 42: 759-769. Cheng, A.L., Hsu, C.H. and Lin, J.K. (2001). Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk pre-malignant lesions. Anticancer Res., 21: 2895-2900. Cordell, G.A. (2002). PC-SPES: a brief overview. Integr Cancer Ther., 1: 271-286. De Feudis, F.V., Papadopoulos, V. and Drieu, K. (2003). Ginkgo biloba extracts and cancer: a research area in its infancy. Fundam Clin Pharmacol., 17: 405-417. Dorai, T., Cao, Y.-C., Dorai, B., Buttyan, R and Katz, A.E. (2001). Therapeutic potential of curcumin in prostate cancer-III: curcumin inhibits proliferation, induces apoptosis and inhibits angiogenesis of LNCaPprostate cancer cells in vivo. Prostate, 47: 293-303. Fassina, G., Vene, R and Morini, M. (2004). Mechanisms of inhibition of tumor angiogenesis and vascular tumor growth by epigallocatechin-3-gallate. Clin Cancer Res., 10: 48654873. Ferrera, N., Carver-Moore, K. and Chen, H. (1996). Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene. Nature, 380: 439-442. Fidler, I.J. (2000a). Angiogenesis and cancer metastasis. Cancer J., 6(suppI2): S134S141. Fidler, I.J. (2000b). Regulation of neoplastic angiogenesis. J Natl Cancer Inst Monographs, 28: 10-14. Folkman, J. (1986). How is blood vessel growth regulated in normal and neoplastic tissue? GHA Clowes Memorial Award Lecture. Cancer Res., 46: 467-473. Folkman, J. (1995). Angiogenesis in cancer, vascular, rheumatoid and other disease. Nature Med., 1: 27-3l. Folkman, J. and Cotran, R (1976), Relation of vascular proliferation to tumor growth. Int Rev Exp Pathol., 16: 207-248. Folkman, J. and Klagsbrun, M. (1987). Angiogenic factors. Science, 235: 442-447. Furness, M.S., Robinson, T.P. and Ehlers, T. (2005). Antiangiogenic agents: studies on fumagillin and curcumin analogs. Curr Pharm Des., 11: 357-373. Gallo, D., Giacomelli, S. and Ferlini, C. (2003). Antitumor activity of the silybinphosphatidylcholine complex, IdB 1016, against human ovarian cancer. Eur J Cancer, 39: 2403-2410. Garcia-Cardena, G. and Folkman, J. (1998). Is there a role for nitric oxide in tumor angiogenesis? J Natl Cancer Inst., 90: 560-56l. Gururaj, A.E., Belakavadi, M., Venkatesh, D.A., Marme, D. and Salimath, B.P. (2002). Molecular mechanisms of antiangiogenic effect of curcumin. Biochem Biophys Res Commun., 297: 934-942. Hahm, E.R, Gho, Y.S. and Park, S. (2004). Synthetic curcumin analogs inhibit activator protein-1 transcription and tumor-induced angiogenesis. Biochem Biophys Res Commun., 321: 337-344. Hanahan, D., Bergers, G. and Bergsland, E. (2000). Less is more, regularly: metronomic dosing of cytotoxic drugs can target tumor angiogenesis in mice. J Clin Invest., 105: 10451047. Hart, I.R, Goode, N.T. and Wilson, RE. (1989). Molecular aspects of the metastatic cascade. Biochim Biophys Acta, 989: 65-84. Hejna, M., Raderer, M. and Zielinski, C.C. (1999). Inhibition of metastases by anticoagulants. J Natl Cancer Inst., 91: 22-26. Hsieh, T.C., Lu, X. and Chea, J. (2002). Prevention and management of prostate cancer using PC-SPES: a scientific perspective. J Nutr., 132(l1suppl): 3513S-35178. Huang, G.W., Xie, C.x. and Kuang, G.Q. (2003). Treatment of 41 patients with advanced stage of nasopharyngeal cancer by combination therapy of radiation and Chinese herbal drugs for activating blood circulation to remove stasis as hirudo. Zhongguo Zhong Xi Yi Jie He Za Ahi, 23: 777-778. Hudis, C.A. (2005). Clinical implications of antiangiogenic therapies. Oncology, 19(4 suppI3): 26-31.
Anti-Angiogenesis Therapies for Treating Cancer
63
Igura, K, Ohta, T., Kuroda, Y. et al., (2001). Resveratrol and quercetin inhibit angiogenesis in mtro. Cancer Lett., 171: 11-16. Ingber, D., Fujita, T. and Kishimoto, S. (1990). Synthetic analogues of fumagillin that inhibit angiogenesis and suppress tumor growth. Nature, 348: 555-557. Jiang, C., Agarwal, Rand Lu, J. (2002). Antiangiogenic potential of a cancer chemopreventive flavonoid antioxidant, silymarin: inhibition of key attributes of vascular endothelial cells and angiogenic cytokine secretion by cancer epithelial cells. Biochem Biophys Res Commun., 276: 371-378. Jin, Y., Zhang, L. and Zhang, M. (2003). Antitumor activities of heteropolysaccharides of Poria cocos mycelia from different strains and culture media. Carbohydr Res., 338: 1517152l. John, V.D., Kuttan, G. and Krishnakutty, K (2002). Anti-tumour studies of metal chelates of synthetic curcuminoids. J Exp Clin Cancer Res., 21: 219-224. Jung, Y.D., Ahmad, S.A. and Akagi, Y. (2000). Role of the tumor microenvironment in mediating response to anti-angiogenic therapy. Cancer Metastasis Rev., 19: 147-157. Khafif, A., Hurst, R, Kyker, K et al., (2005). Curcumin: a new radiosensitizer of squamous cell carcinoma cells from Otolaryngol Head Neck Surg., 132: 317-32l. Kim, E.C., Min, J.K and Kim, T.Y. (2005). [61-Gingerol, a pungent ingredient of ginger, inhibits angiogenesis in VLtro and in vivo. Biochem Bwphys Res Commun., 335: 300-308. Kim, J.H., Shim, J.S. and Lee, S.K (2002). Microarray-based analysis of anti-angiogenic activity of demethoxycurcumin on human umbilical vein endothelial cells: crucial involvement of the down-regulation of matrix metalloproteinase. Jpn J Cancer Res., 93: 1378-1385. Kojima-Yuasa, A., Hua, J.J. and Kennedy, D.O. (2003). Green tea extract inhibits angiogenesis of human umbilical vein endothelial cells through reduction of expression of VEGF receptors. Life Sci., 73: 1299-1313. Kruger, E.A., Duray, P.H., Price, D.K, Pluda, J.M. and Figg, W.D. (2001). Approaches to preclinical screening of antiangiogenic agents. Semin Oncol., 28: 570-576. Kumar, R, Yoneda, J., Bucana, C.D. and Fidler, I.J. (1998). Regulation of distinct steps of angiogenesis by different angiogenic molecules. Int J Oncol., 12: 749-757. Lee, B.C., Doo, H.K and Lee, H.J. (2004). The inhibitory effects of aqueous extract of Magnolia officinalis on human mesangial cell proliferation by regulation of plateletderived growth factor-BB and transforming growth factor beta1 expression. J Pharmacol Sci., 94: 81-85. Lee, K.Y., You, H.J. and Jeong, H.G. (2004). Polysaccharide isolated from Poria cocos sclerotium induces NF-kappaBlRel activation and iNOS expression through the activation of p38 kinase in murine macrophages. Int Immunopharmaco., 4: 1029- 1038. Lee, M.J., Maliakal, P. and Chen, L. (2002). Pharmacokinetics of tea catechins after ingestion of green tea and (-)-epigallocatechin-3-gallate by humans: formation of different metabolites and individual variability. Cancer Epidemiol Biomarkers Prev., 11: 10251032. Leu, T.H., Su, S.L., Chuang, Y.C. and Maa, M.C. (2003). Direct inhibitory effect of curcumin on src and focal adhesion kinase activity. Biochem Pharmacol., 66: 2323-233l. Lin, M.T., Yen, M.L. and Lin, C.Y. (2003). Inhibition of vascular endothelial growth factorinduced angiogenesis by resveratrol through interruption of Src dependent vascular endothelial cadherin tyrosine phosphorylation. Mol Pharmacol., 64: 1029-1036. Liotta, L.A. and Stetler-Stevenson, W.G. (1991). Tumor invasion and metastasis: an imbalance of positive and negative regulation. Cancer Res., 51: 5054-5059. Liu, J.J., Huang, T.S., Cheng, W.F. and Lu, F.J. (2003). Baicalein and baicalin are potent inhibitors of angiogenesis: inhibition of endothelial cell proliferation, migration and differentiation. Int J Cancer, 106: 559-565. Maraveyas, A., Lam, T., Hetherington, J.W. and Greenman, J. (2005). Can a rational design for metronomic chemotherapy dosing be devised? Br J Cancer, 92: 1588-1590. Meade-Tollin, L.C., Wijeratne, E.M. and Cooper, D. (2004). Ponicidin and oridonin are responsible for the antiangiogenic activity of Rabdosia rubescens, a constituent of the herbal supplement PC SPES. J Nat Prod., 67: 2-4.
64
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Miller, KD., Sweeney, C.J. and Sledge, G.W. (2001). Redefining the target: chemotherapeutics as antiantiogenics. J Clin Oncol., 19: 1195-1206. Miocinovic, R, McCabe, N.P. and Keck, RW. (2005). In vivo and in vitro effect of baicalein on human prostate cancer cells. Int J Oncol., 26: 241-246. Mizushina, Y., Akihisa, T. and Ukiya, M. (2004). A novel DNA topoisomerase inhibitor: dehydroebriconic acid; one of the lanostane-type triterpene acids from Poria cocos. Cancer Sci., 95: 354-360. Mueller, M.S., Runyambo, N. and Wagner, I. (2004). Randomized controlled trial of a traditional preparation of Artemisia annua L. (Annual Wormwood) in the treatment of malaria. Trans R Soc Trop Med Hyg., 98: 318-321. Nagy, J.A., Brown, L.F. and Senger, D.R (1989). Pathogenesis of tumor stroma generation: a critical role for leaky blood vessels and fibrin deposition. Bioch~m Biophys Acta, 948: 305-326. Narayan, S. (2004). Curcumin, a multi-functional chemopreventive agent, blocks growth of colon cancer cells by targeting betacatenin-mediated transactivation and cell-cell adhesion pathways. J Mol H~stol., 35: 301-307. Neeman, M. (2000). Preclinical MRI experience in imaging angiogenesis. Cancer Metastasis Rev., 19: 39-43. Oh, W.K, George, D.J. and Hackmann, K (2001). Activity of the herbal combination, PCSPES, in the treatment of patients with androgen-independent prostate cancer. Urology, 57: 122-126. Oh, W.K, Kantoff, P.W. and Weinberg, V. (2004). Prospective, multicenter randomized phase II trial of the herbal supplement: PC-SPES, and diethylstibestrol in patients with androgen- independent prostate cancer. J Clin Oncol., 22: 3705-3712. Oliff, A., Gibbs, J.B. and McCormick, F. (1996). New molecular targets for cancer therapy. Sci Am., 275: 144-149. Pisters, KM.W., Newman, RA. and Coldman, B. (2001). Phase I trial of oral green tea extract in adult patients with solid tumors. J Clin Oncol., 19: 1830-1838. Pyun, H.J., Fardis, M. and Tario, J. (2004). Investigation of novel fumagillin analogues as angiogenesis inhibitors. Bworg Med Chem Lett., 14: 91-94. Reddy, S. and Aggarwal, B.B. (1994). Curcumin is a non-competitive and selective inhibitor of phosphorylase kinase. Fed Eur Bwchem Soc Lett., 341: 19-22. Ria, R, Portaluri, M. and Russo, F. (2004). Serum levels of angiogenic cytokines decrease after antineoplastic radiotherapy. Cancer Lett., 216: 103-107. Risau, W. (1997). Mechanisms of angiogenesis. Nature, 386: 671-674. Ruegg, C., Meuwly, J.-Y. and Driscoll, R. (2003). The quest for surrogate markers of angiogenesis: a paradigm for translational research in tumor angiogenesis and anti angiogenesis trials. Curr Mol Med., 3: 673-691. Sagar, S.M. (2007). Future directions for research on Silybum marianum for cancer patients. Integrat Cancer Ther., 6: 166-173. Salcedo, X., Medina, J. and Sanz-Cameno, P. (2005). Review article: angiogenesis soluble factors as liver disease markers. Al~ment Pharmacol Ther., 22: 23-30. Samuels, N. (2005). Herbal remedies and anticoagulant therapy. Thromb Haemost., 93: 3-7. Sartippour, M.R, Shao, Z.M. and Heber, D. (2002). Green tea inhibits vascular endothelial growth factor (VEGF) induction in human breast cancer cells. J Nutr., 132: 2307-2311. Sato, K, Mochizuki, M. and Saiki, I. (1994). Inhibition of tumor angiogenesis and metastasis by a saponin of Panax ginseng, ginsenoside-Rb2. Bioi Pharm Bull., 17: 635-639. Schneider, M., Tjwa, M. and Carmelie, P. (2005). A surrogate marker to monitor angiogenesis at last. Cancer Cell., 7: 3-4. Sen, S., Sharma, H. and Singh, N. (2005). Curcumin enhances vinorelbine mediated apoptosis in NSCLC cells by the mitochondrial pathway. Biochem Biophys Res Commun., 331: 1245-1252. Senger, D.R., Galli, S.J. and Dvorak, A.M. (1983). Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science, 219: 983-985. Shaked, Y., Bertolini, F. and Man, S. (2005). Genetic heterogeneity of the vasculogenic phenotype parallels angiogenesis: implications for cellular surrogate marker analysis of antiangiogenesis. Cancer Cell, 7: 101-111.
Anti-Angiogenesis Therapies for Treating Cancer
65
Shao, Z.M., Shen, Z.Z. and Liu, C.H. (2002). Curcumin exerts multiple suppressive effects on human breast carcinoma cells. Int J Cancer, 98: 234-240. Shim, J.S., Kim, J.H. and Cho, H.Y. (2003). Irreversible inhibition of CD13/aminopeptidaseN by the antiangiogenic agent curcumin. Chem Bioi., 10: 695-704. Singh, N.P. and Lai, H.C. (2004). Artemisinin induces apoptosis in human cancer cells. Ant~cancer Res., 24: 2277-2280. Singh, R.P. and Agarwal, R. (2003). Tumor angiogenesis: a potential target in cancer control by phytochemicals. Curr Cancer Drug Targets, 3: 205-217. Singh, R.P., Sharma, G., Dhanalakshmi, S., Agarwal, C. and Agarwal, R. (2003). Suppression of advanced human prostate tumor growth in athymic mice by silibinin feeding is associated with reduced cell proliferation, increased apoptosis, and inhibition of angiogenesis. Cancer Ep~demiol Biomarkers Prev., 12: 933- 939. Small, E.J., Frohlich, M.W. and Bok, R. (2000). Prospective trial of the herbal supplement PC-SPES in patients with progressive prostate cancer. J Clin Oncol., 18: 3595-3603. Smorenburg, S.N. and Van Noorden, C.J.F. (2001). The complex effects ofheparins on cancer progression and metastasis in experimental studies. Pharmacol Rev., 53: 93-105. Son, H.J., Lee, H.J. and Yun-Choi, H.S. (2000). Inhibitors of nitric oxide synthesis and TNFa expression from Magnolia obovata in activated macrophages. Planta Med., 66: 469471. Sreejayan, R. (1997). Nitric oxide scavenging by curcuminoids. J Pharm Pharmacol., 49: 105-107. Sugarbaker, E.V. (1979). Cancer metastasis: a product oftumor-host interactions. Curr Probl Cancer, 3: 1-59. Sweeney, C.J., Miller, K.D. and Sledge, G.W. (2003). Resistance in the antiangiogenic era: nay-saying or a word of caution? Trends Mol Med., 9: 24-29. Tang, F.Y. and Meydani, M. (2001). Green tea catechins and vitamin E inhibit angiogenesis of human microvascular endothelial cells through suppression of Il-8 production. Nutr Cancer, 41: 119-125. Tang, F.Y., Nguyen, N. and Meydani, M. (2003). Green tea catechins inhibit VEGF-induced angiogenesis in vitro through suppression ofVE-cadherin phosphorylation and inactivation of Akt molecule. Int J Cancer, 106: 871-878. Taraboletti, G. and Giavazzi, R. (2004). Modelling approaches for angiogenesis. Eur J Cancer, 40: 881-889. Tseng, S.H., Lin, S.M. and Chen, J.C. (2004). Resveratrol suppresses the angiogenesis and tumor growth of gliomas in rats. Clin Cancer Res., 10: 2190-2192. Wallace, J.M. (2002). Nutritional and botanical modulation of the inflammatory cascade: eicosanoids, cyclooxygenases, and lipoxygenases as an adjunct in cancer therapy. Integr Cancer Ther., 1: 7-37 Wang, S., Zheng, Z. and Weng, Y. (2004). Angiogenesis and antiangiogenesis activity of Chinese medicinal herbal extracts. Life Sci., 74: 2467-2478. Xu, G.Z., Cai, W.M. and Qin, D.X. (1989). Chinese herb "destagnation" series I: combination of radiation with destagnation in the treatment of nasopharyngeal carcinoma-a prospective randomized trial on 188 cases. Int J Radiat Oncol Bioi Phys., 16: 297-300. Yoshida, S., Ono, M. and Shono, T. (1997). Involvement ofinterleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor-alpha dependent angiogenesis. Mol Cell Bioi., 17: 4015-4023. Zhang, L., Rui, Y.C. and Yang, P.Y. (2002). Inhibitory effects of Ginkgo biloba extract on vascular endothelial growth factor in rat aortic endothelial cells. Acta Pharmacol Sin., 23: 919-923.
"This page is Intentionally Left Blank"
4 An Anthology of the Studies on the AntiInflammatory, Antinociceptive and Febrifuge Effects of Medicinal Plants in Turkey ESRA KUPELI AKKOL 1 AND ERDEM YESlLADA2
ABSTRACT Inflammatory diseases are among the most common health problems treated with traditional remedies. Therefore it is crucial to evaluate the potential of herbal remedies for the discovery of novel bioactive compounds that might serve as leads for the development of potent drugs. Although several agents are known to treat these types of disorders, prolonged use should be avoided due to severe side effects. Consequently, there is a need to develop safe and new anti-inflammatory agents with minimum side effects. In this context, traditional medicines provide a vast source for the discovery of original drug leads. The present investigation represents a preliminary screening in an ongoing program on plants used in Turkish traditional medicine for the treatment of rheumatism and related inflammatory diseases. In this study, selected plants as the subject of the present study were recorded in the alphabetical order of plant families. Key words : Antinociceptive, anti-inflammatory, bioassay-guided processing, ethnopharmacology, febrifuge, folk remedies, plants, Turkey 1. Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Etiler 06330, Ankara, Turkey. 2. Department of Pharmacognosy, Faculty of Pharmacy, Yeditepe University, Kayisdagi 34755 Istanbul-Turkey. * Corresponding author: E-mail:
[email protected]
68
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
INTRODUCTION Acute and chronic inflammations are known to be complicated processes induced by several different classes of chemical mediators, e.g. prostaglandins, leukotrienes and platelet-activating factor, etc. Antiinflammatory agents exert their effect through a spectrum of different modes of action (Samuelsson et al., 1978). However, chronic inflammatory diseases are still one of the major health problems worldwide and nonsteroid anti-inflammatory drugs (NSAIDs) are the most prescribed drugs for treatment of inflammatory diseases. Although the NSAIDs provide the patients with symptomatic relief, they do not modify the pathogenesis of inflammation and do not reduce the disabling bone and cartilage damage (Ford-Hutchinson et al., 1981). Therefore, it has become a must to search for new initiatives in the treatment of chronic inflammation. On the other hand, plants have been used by human being since ages in traditional medicine due to their therapeutic potential and the search on medicinal plants have led the discovery of novel drug candidates used against diverse diseases. The plant kingdom encompasses a wide range of plants that possess biologically active molecules of medicinal value. Many such plant-derived molecules have been isolated, identified, and successfully introduced into international markets by pharmaceutical industries that are mostly located in the developed world. In contrast, plant-based remedies, prescribed by folk doctors in the developing world for the treatment of many diseases, remain largely restricted in their use to the local communities in which such remedies were developed (Sumner, 2000). Although such plant-based remedies have recently received considerable interest from the medical and pharmaceutical industries, only a small fraction of such plants have been assessed. In the past few years, many studies have investigated the healing potential of higher plants with ethnobotanical histories. This stemmed from several reasons such as consumer interest in such plantbased remedies, the concern about the possible side effects of allopathic medicine, the lower cost of phytotherapy, and the fact that many plantbased or herbal remedies succeeded in replacing allopathic medicines in relieving malady symptoms. The aim of this manuscript is to review the literature of bioactive anti-inflammatory plant extracts and their isolated active components as an addition to the literature typically used in folk medicine in Turkey basin to treat inflammation related ailments.
ANACARDIACEAE The nuts of Pistacia vera L. (Anacardiaceae), commonly referred to pistachio, is a quite popular flavoring foodstuff and snack in Turkey and in the world. The oleoresin of P. vera, a widely-distributed tree throughout
Effects of Medicinal Plants in Turkey
69
the south-east region of Anatolia, has been used to treat asthma by chewing as folk remedy in this part of Turkey as well as against stomachache (by chewing the gum) and hemorrhoids (externally) (Baytop, 1999). In the light oftraditional information, the ethanolic and aqueous extracts prepared from different parts (leaf, branch, fruit, stalk) of Pistacia vera as well as its oleoresin were evaluated for their in vivo anti-inflammatory and antinociceptive activities (Orhan et al., 2006). Only the oleoresin, out of the all extracts screened herein, exhibited a dose-dependent anti-inflammatory activity on carrageenan-induced hind paw edema model in mice ranging between 27.4-34.7% at 250 mg/kg and 32.6-38.7% at 500 mg/kg doses without inducing any gastric damage. The potency of oleoresin at 500 mg/kg dose was almost equal to that observed for indomethacin, a reference agent. As for antinociceptive activity, the oleoresin also displayed 32.1 % inhibition at 500 mg/kg dose onp-benzoquinone induced abdominal contractions in mice, while inhibitory rate decreased to 21.7% at 250 mg/kg. Fractionation of the oleoresin indicated that n-hexane fraction to be active, which further led to recognition of some monoterpenes, mainly a-pinene (77.5%) by capillary gas chromatography-mass spectrometry (GC-MS) as well as the oleoresin itself. a-Pinene was also assessed for its antinociceptive and antiinflammatory activities in the same manner and exerted a moderate antiinflammatory effect at 500 mg/kg dose. APIACEAE In Turkish folk medicine various species of the Eryngium are used for a wide range of ailments; particularly roots are used against various inflammatory disorders, oedema, sinusitis, urinary infections or inflammations etc. and snake or scorpion bites or goiter, roots and leaves for infertility and herbs for wound healing as well as food while fresh (Sezik et al., 1997; Yesilada & Sezik, 2003). Eryngium species, particularly E. campestre and E. maritimum are called in Turkish folk-medicine as "Bogadikeni" and widely distrubuted in all parts of Turkey. Infusion of aerial parts and roots of these species are used in folk remedies as antitussive, diuretic, appetizer, stimulant and aphrodisiac (Baytop, 1999). Aqueous extract ofthe roots of E. campestre is used orally and poultice is applied externally as a remedy aganst scorpion bite in the Balikesir-Turkey (personal interview). Similar utilizations were found in the old documents from the medieval ages such as against wound and burns, pains, hemorrhoids and sexual diseases (Lev, 2002). Effects of the roots from different Eryngium species against various inflammatory conditions are also well-known in the traditional medicines worldwide; for example, E. campestre as anti-oedema (Leporatti & Ivancheva, 2003) or E. creticum for inflammatory wounds (AliShtayeh et al., 1998). It is also documented in the popular phytotherapy books for such treatments, i.e. E. campestre is recommended against kidney and urinary tract inflammations and oedema (Gruenwald et al. 2000a). To
70
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
confirm the traditional usage ethanolic and aqueous extracts obtained from either aerial parts or roots of E. campestre, E. creticum, E. davisii, E. falcatum, E. isairicum, E. kotschyi, E. maritimum, and E. trisectum were evaluated for their in vivo anti-inflammatory and antinociceptive activities (Kiipeli et al., 2006a). According to the results of investigations, except E. falcatum extracts, ethanol extracts either from the aerial parts or roots of Eryngium species showed apparent anti-inflammatory activity on carrageenan-induced hind paw oedema and TPA-induced ear oedema tests and antinociceptive activity on p-benzoquinone-induced writhing test. In spite of potent activity of the ethanol extract from E. isairicum aerial parts was induced gastric damage. Aerial parts and roots of E. maritimum and E. kotschyii were found to possess most promising activities without including any apparent gastric damage. Moreover, Yesilada et al. (1989) reported that the methanol extract (150 mg/kg) from the whole plant of E. billardieri showed a 44% inhibition on increased vascular permeability induced by acetic acid in mice on oral administration. Further studies were conducted on the various solvent fractions obtained from the aerial parts and roots of the plants and they were tested for their effects on increased vascular permeability induced by acetic acid model, as well as serotonin, bradykinin, and carrageenan-induced paw oedemas in mice. A significant antiinflammatory activity was observed only in the precipitated part of the nbutanol extract of the roots, which mainly contains saponins (Yesilada et al., 1989).
Seseli is an old Greek name that was called by Hippocrates for certain members of the Apiaceae family (Hamlyn, 1969). The genus Seseli L., is composed of aromatic herbs and economically important species that are used as foods, spices, condiments and ornamentals (Lawrence, 1969; Crowden et al., 1969; Pimenov & Leonov, 1993). Several Seseli species are reported in ancient literature for various healing effects. The roots of S. mairei Wolff., have been used against inflammations, swelling, rheumatism, pain and common cold in Chinese traditional medicine (Hu et al., 1990). The seeds of an Indian species, S. indicum, have been reported to possess anthelmintic, carminative, stomachic and stimulant properties (Tandan et al., 1990). In Turkish folk medicine, the fruit of S. tortuosum is used as emmenagogue and anti-flatulence (Baytop, 1999), while the leaves of S. libanotis (Kelemkesir or Kelemenkesir in Turkish) are consumed as a vegetable in the eastern Turkey (Baytop, 1999). Traditional usage of the genus in folk medicine led us to search other species for their biological activies. The ethyl acetate and methanol (80%) extracts obtained from ten Seseli L. species (Apiaceae) growing in Turkey, Seseli andronakii Woron., S. campestre Besser, S. gummiferum Pall. ex Sm. subsp. corymbosum (Boiss & Heldr) P. H. Davis, S. gummiferum Pall. ex Sm. subsp. gummiferum, S. hartvigii Parolly & Nordt, S. libanotis (L.) W. Koch, S. petraeum M. Bieb., S. peucedanoides (Bieb.) Koso-Pol., S. resinosum Freyn & Sint., S. tortuosum
Effects of Medicinal Plants in Turkey
71
L. were evaluated for their in vivo anti-inflammatory and antinociceptive activities (Kiipeli et al., 2006c). Among the plant extracts, the ethyl acetate extracts from S. andronakii, S. campestre, S. gummiferum subsp. corymbosum, S. petraeum, S. resinosum and S. tortuosum showed 30.1%, 32.3%, 36.9%, 39.8%, 35.1%, 37.6% inhibition in p-benzoquinone-induced abdominal constriction test, respectively. The ethyl acetate extracts of S. gummiferum subsp. corymbosum, S. petraeum, S. resinosum and S. tortuosum also exhibited notable inhibition, ranging between 24.5-29.7%, 28.1-33.3%, 17.4-27.5%, 27.9-31.3%, respectively, in carrageenan-induced hind paw edema model at 100 mg/kg dose without inducing any gastric damage. During the acute toxicity evaluation, neither death nor gastric bleeding was observed for any of the plant extracts. Results have supported the traditional use of some Seseli species against inflammatory disorders. APOCYNACEAE
Nerium oleander L., oleander, is widely distributed in the Mediterranean region. Due to the toxic nature, limited number of utilization have been recorded in Turkish folk medicine. Among them a speculative assertion on cancer treatment by a Turkish medical doctor became very popular and eventuated recently the leaf extract undergoing Phase I clinical evaluation as a potential treatment against cancer in USA (Smith et al., 2001). During our expeditions, an informant described that fresh oleander flowers was put in alcohol in summer and kept as a home-remedy to alleviate her severe pain and paralysis in her legs. In a reference survey in the database program of Turkish folk medicine (TUHIB), a similar utilization was also encountered by another informant, i.e. flowers kept 40 days in olive oil was applied on joint against rheumatic pain (Yesilada, 2002). Moreover, leaves or flowers are used to stop pain or eczema, sap obtained from fresh leaves for abscess or rheumatism were recorded. Besides, another part of oleander, the fruits, is reported as antirheumatic and as a remedy for skin diseases in Saudi folk medicine (Adam et al., 2001). In this context, Zia et al. (1995) reported that two purified fractions (B 1 and B3 ) obtained from the methanol extract of fresh oleander leaves possess a eNS depressant activity, i.e. produced reduction in locomotor activity, rotarod performance and potentiate hexobarbital induced sleeping time. Moreover, they showed significant analgesic activity as indicated by inhibitory effects on acetic acidinduced writhings and increased reaction time to thermal test. Our results also supported that both aqueous and ethanol extracts of oleander leaves possess significant antinociceptive activity as decided by using another writhing test model, but the effect of EtOH extract was more pronounced (Erdemoglu et al., 2003). However, both extracts were shown gastric ulcerogenicity in mice. On the other hand, EtOH extracts from either dried or fresh flowers exhibited potent antinociceptive activity without inducing any gastric damage and potency was more pronounced than that of reference
72
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
drug aspirin. Experimental results have clearly confirmed that liposoluble fractions of flowers, as decided by ethanol or olive oil extracts were used in folk medicine, possess remarkable antinociceptive activity. Antiinflammatory activity assessment experiment has also verified the conclusion that liposoluble components of flowers may have the activity since only the EtOH extracts were found active even than that of reference drug indomethacin.
ASTERACEAE The genus Achillea, named after the mythological Greek warrior Achilles, comprises of approximately 85 species, most of which are endemic to Europe and the Middle East. In the Turkish flora 40 Achillea species have been recorded and 20 of them are endemic (Davis, 1982). On the otherhand, seveal Achillea species have been known to be used in folk remedies for various purposes such as haemorrhoid and wound healing (Baytop, 1999). Especially, A. millefolium is frequently used against diarrhoea, abdominal pain and stomachache in Turkish traditional medicine (Yesilada et al., 1993; Fujita et al., 1995; Honda et al., 1996). In order to evaluate their folkloric utilization, both antinociceptive and anti-inflammatory activities of five Achillea species including Achillea wilhelmsii C. Koch,A. setacea Waldst & Kit, A. vermicularis Trin., A. phrygia Boiss. & Bal. and A. sipikorensis Hausskn. et Bornm. were investigated (Kiipeli et al., 2007a). The ethanol extracts of A. wilhelmsii, A. setacea and A. vermicularis showed significant antinociceptive (p-benzoquinone-induced writhing test) and antiinflammatory (carrageenan-induced hind paw edema model) activity at 500 mg/kg dose, per os, without inducing any apparent acute toxicity as well as gastric damage. A. phrygia was shown to possess only significant antinociceptive activity, while A. sipikorensis did not show any remarkable anti-inflammatory and antinociceptive activity. Furthermore, the essential oils obtained by water distillation from aerial parts of Achillea schischkinii Sosn. and Achillea aleppica DC. subsp. aleppica were analysed by GC and GC/MS. 1,8-Cineole (32.5% and 26.1%, respectively) was the main component in both oils. Their antimicrobial, anti-inflammatory and antinociceptive activities were also studied and the oil of A. aleppica subsp. aleppica showed significant anti-inflammatory, antinociceptive and moderate antimicrobial activities. The oil of Achillea aleppica subsp. aleppica contains 6.6% of a-bisabolol and its derivatives. These compounds might be at least partially responsible for antiinflammatory activity of the oil (Iscan et al., 2006). In folk medicine, Helichrysum species have been used for gall bladder disorders as medicinal tea, because of their bile regulatory and diuretic effects (Siizge~ et al., 2005). Some of them are used for anti-inflammatory and anti-allergic properties (Carini et al., 2001; Tepe et al., 2005). Moreover,
Effects of Medicinal Plants in Turkey
73
this genus is traditionally used in the treatment of wounds, infections and respiratory conditions (Lourens et al., 2004). In Turkey, several Helichrysum species are used in folk medicine as diuretic, cholagogue, and against kidney stones (Baytop, 1999). In order to evaluate the anti-inflammatory and antinociceptive activities of Helichrysum species (Kiipeli et al., 2006b), aqueous extracts from H. arenarium (L.) Moench. subsp. aucheri (Boiss.) Davis & Kupicha and H. plica tum DC. subsp. plicatum were investigated and they showed 37.0%, 30.4% inhibition at 100 mg/kg doses and 31.2%, 28.3% at 200 mg/kg doses in p-benzoquinone-induced abdominal constriction test, respectively, without inducing any gastric damage. The aqueous extracts of all the plants did not show any anti-inflammatory activity in carrageenaninduced hind paw edema model at 50, 100 and 200 mg/kg doses. During the acute toxicity evaluation, neither death nor gastric bleeding was observed for any of the plant extracts.
BERBERIDACEAE Roots and barks of various Berberis species are used as folk remedy for the treatment of various inflammatory diseases such as lumbago, rheumatism and to reduce fever. In Semerkand Bazaar (Uzbekistan) a black tough material prescribed for the treatment of lumbago attracted our attention during our scientific expeditions. The herb dealer described that it was the concentrated aqueous extract of Berberis obionga roots. In a reference survey, several other Berberis species were also found to be used in the treatment of various inflammatory conditions, including rheumatism, fever and pyrexia. In order to evaluate this information, the effect of a widespread Turkish species, B. crataegina DC. was studied and found that the roots of the plant possessed significant anti-inflammatory, analgesic and febrifuge effects. Through bioassay-guided fractionation and isolation procedures berberine was isolated as the main active ingredient responsible from the relevant effects. In that paper, however, it was addressed that the role of other alkaloids in the effect of the roots should also be investigated (Yesilada & Kiipeli, 2002). In the study of Kosar et al. (1999) three types ofisoquinoline alkaloids were detected in the roots, barks and branches of Turkish Berberis species: protoberberine (berberine, palmatine,jatrorrhizine, columbamine), bisbenzylisoquinoline (berbamine, oxyacanthine, aromoline) and aporphine (magnoflorine) types. In a following study, the effects of these alkaloids were studied using various in vivo models in mice (Kiipeli et al., 2002). All alkaloids inhibited inflammations in varying degrees, among them berberine, berbamine and palmatine were shown to possess significant and dosedependent inhibitory activity against serotonin-induced hind paw oedema both on oral and topical applications and acetic acid-induced increase in vascular permeability on oral administration. Moreover, these three alkaloids were also shown to possess dose-dependent antinociceptive activity,
74
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
which assessed by using the model based on the inhibition of p-benzoquinoneinduced writhing movements as well as antipyretic activity on FCA-induced increased rectal temperature on subacute administration. However, all alkaloids induced gastric lesions in varying degrees (Kiipeli et al., 2002). Since it is well known that anti-inflammatory agents acting through inhibition of cyclooxygenase-l (COX-I) enzyme induce such gastric ulceration, the effect of isoquinoline alkaloids might also be based on this mechanism. As a matter of fact, berberine was found to inhibit castor oilinduced diarrhoea in rats, which supported this conclusion (Yesilada & Kiipeli, 2002). Fukuda et al. (1999) demonstrated that berberine inhibits cyclooxygenase-2 (COX-2) transcriptional activity in colon cancer cells in a dose-dependent manner, however, the effect on constitutive enzyme (COX1) was not determined. It is noteworthy that powdered roots of B. lycium is suggested to be taken with milk for the treatment of rheumatism and muscular pains in Pakistan folk medicine, probably to protect the gastric mucosa from damage (Ikram et al., 1966). The results of the previous study (Kiipeli et al., 2002) revealed that berberine, berbamine and palmatine were the main anti-inflammatory, antinociceptive and antipyretic alkaloids in the roots of Berberis species. Especially the effect of first two was more pronounced. Since alkaloid composition ofthe roots showed great variations from species to species, the relevant activity would highly be species-specific. Thus, utilization of B. integerrima roots in therapy would not be beneficial due to the higher concentrations of the alkaloids with weak activity, i.e. magnoflorine and oxyacanthine. On the other hand, due to the higher concentration of effective alkaloids in B. cretica and B. vulgaris roots, more beneficial effects would be expected. Berberine and related isoquinoline alkaloids have been reported to exhibit a broad distribution in the plant kingdom (Keys, 1985), such as; Epimedium sp. leaves (Berberidaceae); Phellodendron sp. barks and Xanthophyllum sp. roots (Rutaceae); Coptis sp. rhizomes and Hydrastis sp. roots (Ranunculaceae); Sanguinaria sp. rhizomes (Papaveraceae); etc. In traditional medicine, some of these plants are known to be employed in the treatment of various inflammatory ailments and results of the present study would also be helpful to explain the relevant effects as well as safety evaluation of these plant remedies especially regarding to the ulcerogenic effect of these alkaloids reported in that study (Kiipeli et al., 2002). Contrary to the observations of the present study that berberine type alkaloids induce remarkable gastric lesions, berberine was reported to be effective in the treatment of gastritis and gastric ulcers in some documents (PDR, 2000), which needs further studies and explanations.
BRASSICACEAE In Turkish folk medicine, Isatis sp. is known to be used for wound healing and against constipation (Baytop, 1999). In Iran, another species of the
Effects of Medicinal Plants in Turkey
75
plant, 1. cappadocia, has also been reported against rheumatism, asthma, eczema, fever, headache and wound healing. Rezaeipoor et al. (2000) showed that the plant possess in vivo suppressive effect on humoral primary immune response at the dose of 250 mg/kg, while stimulated secondary immune responses at the dose of 500 mg/kg. Additionally, the activity of a Turkish species, Isatis glauca Aucherx Boiss. ssp. glauca, was studied and ethanol extract demonstrated a significant antinociceptive activity (37.2%) at 500 mg/kg, and a medium anti-inflammatory activity (24-28% inhibition) (Kupeli et al., 2007c).
CISTACEAE In Turkish folk medicine, the leaves of the Cistus laurifolius are also used externally as an effective remedy against several inflammatory ailments such as rheumatic pain, high fever and urinary inflammations. For relieving rheumatic pain a warm decoction of leaves is used as a bath or wilted leaves are externally applied onjoints. For alleviation or treating of urinary inflammations, a poultice is prepared by boiling the leaves and then mixing with flour. This poultice is then applied externally as a plaster on the dorsal part ofthe body in a line with the kidneys (Sezik et al., 1991; Yesilada et al., 1995). Effects of the extracts and fractions from the leaves with non-woody branches ofCistus laurifolius L. (Cistaceae) were studied using two in vivo models of inflammation in mice. For the preliminary antiinflammatory activity assessment of C. laurifolius leaves two types of extracts (aqueous and ethanolic) were prepared and investigated against carrageenan-induced hind paw oedema and acetic acid-induced increased vascular permeability models in mice. Aqueous extract did not show any remarkable effect against carrageenan-induced hind paw edema model, while EtOH extract was significantly active both in 250 and 500 mg/kg doses. A comparable result was obtained against another inflammatory model based on the inhibition of increased vascular permeability, induced by the intraperitoneally injection of acetic acid. Subsequently, through bioassay-guided fractionation and isolation procedures three flavonoids; 3-0-methylquercetin, 3,7 -O-dimethylquercetin and 3,7-0dimethylkaempferol were isolated as the main active ingredients from the ethanol extract (Fig 1). Moreover, these flavonoids were shown to possess potent antinociceptive activity, which was assessed through inhibition ofp-benzoquinone-induced writhing reflex. Results ofthe present study have clearly supported the utilization of C. laurifolius in Turkish traditional medicine and three flavonoids were shown to have strong antinociceptive and anti-inflammatory activities, per os without inducing any apparent acute toxicity as well as gastric damage (Kiipeli & Yesilada, 2007).
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
76
OH Me O
OH
OH
OH
OH
o
0
OH MeO
OH
0
Fig 1. Chemical structures of active compounds
DIOSCOREACEAE Tamus communis root is used to treat rheumatism after pounded in Turkish folk medicine (Yesilada et al., 1995). In a previous report, the ethanol extract from Tamus communis roots was shown to possess significant inhibitory effect on granuloma formation when tested on cotton pellet granuloma model in rats (Capasso et al., 1983; Mascolo et al., 1987). However, in our study neither aqueous nor ethanol extracts prepared from both roots and aerial parts of that plant did show any remarkable anti-inflammatory or antinociceptive activity (Kupeli et al., 2007 c).
ERICACEAE The sap obtained from a fresh branch of Rhododendron ponticum L. is dropped into the toothcave against toothache in Turkish folk medicine (Yesilada et al., 1999). The flowers and fruits of another Rhododendron species, R. molle, have also been recorded in ancient and modern monographs as analgesic and insecticide in Chinese Traditional Medicine (Li et al., 2000). Moreover, the leaves of R. chrysanthum in Siberia (Grieve, 1994) and R. ferrugineum in Germany (Chosson et al., 1998) have been reported as a remedy against rheumatism. In a reference survey, we did not find any experimental study related to this subject, however, Manjeet and Ghosh (1999) reported that quercetin isolated as a major component of R. cinnabarium inhibited LPS-induced nitric oxide and tumor necrosis factor-production in murine macrophages, which might be related with the aforementioned effects. In vivo studies also confirmed that leaves of the
Effects of Medicinal Plants in Turkey
77
plant possess a significant antinociceptive activity. The EtOH extract showed the highest antinociceptive activity among the plants investigated as well as a potent anti-inflammatory activity without inducing any gastric lesion was recorded (Erdemoglu et al., 2003). The genus Erica is represented only four species of which one taxon (E. bocquetti (Pe~men) P. F. Stevens) is endemic in Turkey. Among the other species, E. arborea L. and E. manipuliflora Salisb. are widespread species common in the coastal sides in Turkey, while E. sicula Guss. subsp. libanotica (C. & W. Barbey) P. F. Stevens has a limited distribution in South West Anatolia (Stevens, 1978). Herbal tea prepared from aerials parts of E. arborea or E. manipuliflora has been popularly used as diuretic, astringent and treatment of urinary infections in Turkey (Baytop, 1999; Tuzlaci & Erya~ar Aymaz, 2001). In particular, a glass of 5% infusion or decoction of E. arborea leaves is suggested after meals to discharge the edema from the body in slimming formulations (Ba~er et al., 1986; Antonone et al., 1988). In the light of traditional usage anti-inflammatory and antinociceptive activities of different extracts were prepared with methanol, chloroform, ethyl acetate, n-butanol and water from the aerial parts of E. arborea L., E. manipuliflora Salisb., E. bocquetii (Pesmen) P. F. Stevens and E. sicula Guss. subsp. libanotica (C. & W. Barbey) P. F. Stevens (Ericaceae) of Turkish origin were investigated by using in vivo methods (Kiipeli Akkol et al., 2008c). The ethyl acetate extracts of E. arborea (EAE), E. bocquetii (EBE) andE. manipuliflora (EME) exhibited notable inhibition against carrageenan-induced (24.1-32.3%, 23.8-36.1%, 29.2-35.1%, respectively) and PGE 2 -induced (21.2-37.7%, 6.8-29.7%, and 6.2-34.1%, respectively) hind paw edema as well as TPA-induced mouse ear edema models in mice, while the ethyl acetate extract of E. sicula subsp. libanotica (ESE) (10.7-29.7%) displayed potent anti-inflammatory activity only on the PGE 2-induced hind paw edema model. However, the remaining extracts were found to be inactive against inflammatory models. Same extracts i.e., EAE, EBE and EME were also found to exhibit remarkable antinociceptive activity inp-benzoquinone-induced abdominal constriction test at a dose of 100 mg/kg (46.5%,27.7% and 36.3%, respectively) (Kiipeli Akkol et al., 2008c). The anti-inflammatory and antinociceptive activities were mainly determined in the ethyl acetate extracts. Previous studies have shown that flavonoids, anthocyanoids, coumarins, terpenic compounds especially ursolic acid and essential oils were present in Erica species (Vieitez & Ballester, 1972; Ballester et al., 1975; Bennini et al., 1993; Chulia et al., 1995; Dufor et al., 2007). Flavonoids exert significant scavenging properties on oxygen radicals and can interfere with the production of arachidonic acid metabolites trough lipoxygenase enzyme inhibition and reduce the concentration of leukotrienes (Reyes Ruiz et al., 1996; Halliwell & Chirica 1993).
78
Camp. Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
Calluna vulgaris L. (heather), which is also known as "the true heather of Europe", is the sole member ofthe genus Calluna (Ericaceae) in Turkey (Baytop, 1999). Heather has a long history of medicinal use in folk medicine. In Anatolia, infusion ofthe plant has been used as urinary disinfectant and diuretic (Baytop, 1999). Different ethnobotanical uses ofthe plant have been documented as antiseptic, astringent, cholagogue, depurative, diaphoretic, diuretic, expectorant, mildly sedative, vasoconstrictor and antirheumatic as well as for the treatment of gout symptoms in European societies (Mabey, 1979; Chiej, 1984; Kumarasamy et al., 2002). A homeopathic remedy is prepared from the fresh branches is prescribed in the treatment of rheumatism, arthritis and insomnia (Launert, 1981). The plant is also suggested among the Bach flower remedies and the keywords for prescribing it are "self-centeredness" and "self-concern" (Chancellor, 1985). In order to evaluate this ethnobotanical information, its anti-inflammatory and antinociceptive activities were studied using in vivo experimental models in mice (Orhan et al., 2007b). The ethanolic extract of the plant was first fractionated into five extracts; namely, n-hexane, chloroform, ethyl acetate (EtOAc), n-butanol, and water fractions. Among them, the EtOAc fraction was found to be the most effective and was further subjected to bioassayguided fractionation and isolation procedures. After successive column chromatography applications, on Sephadex LH-20 and silica gel, a component, which is responsible for the above-mentioned activities ofthis species of Turkish origin, was isolated and its structure was elucidated as kaempferol-3-0-[3-D-galactoside, a common flavonol derivative by means of spectral techniques (Fig 2) (Orhan et al., 2007b). A reference survey has revealed that in vitro anti-inflammatory activity of C. vulgaris was previously OH HO
o OH
o
;::O~~
'~ HO
Fig 2. Chemical structure ofkaempferol-3-0-j3-D-galactoside
reported by two separate studies. In the first study, the water extracts of 52 plants from 28 different families, where selections were based upon the records for their use in inflammatory diseases and treatment of wounds in Swedish traditional medicine, were investigated for their inhibitory activities on prostaglandin biosynthesis and platelet activating factor (PAF)-induced exocytosis in vitro and was found highly efficient on both assays (Tunon et
Effects of Medicinal Plants in Turkey
79
al., 1995). In particular, C. vulgaris was one of the three plants, which exhibited the most potent cyclooxygenase inhibition in that study. In connection to these data, the flowers of C. vulgaris, was tested in vitro for its effect on arachidonic acid metabolism using mouse peritoneal macrophages, human platelets, and differentiated HL60 leukemic cells (N ajid et al., 1992). Among the several organic and aqueous extracts of heather flowers, the acetone extract was found to inhibit lipoxygenase (LOX) enzyme and further analysis of this extract led to the isolation of ursolic acid as an active anti-lipoxygenase component. Ursolic acid was shown a higher efficiency on 5-LOX inhibition than 12- and 15-LOX, whereas it had a 38% inhibition on cyclooxygenase. Many flavonoids, isolated from medicinal herbs used in traditional medicines, are endowed with desired biological activities su~h as antimicrobial, anti-hepatotoxic, anti-inflammatory, cardioprotective, antiosteoporotic, and anti-cancer (Middleton, 1998). Amongst them, quercetin and kaempferol are known to be the most common flavonols present in many plants in different glycosidic forms. In one study, two coumaryl glycosides ofkaempferol (platanoside and tiliroside) were reported to exhibit strong anti proliferative effect suppressing DNA synthesis in a dose- and time-dependant manner (Dimas et al., 2000). In a very good accordance with the above data (Orhan et al., 2007b), Gil et al. (1994) also figured out that kaempferol-3-0-f3-D-galactoside possessed a remarkable antiinflammatory activity on both in vivo carrageenan-induced hind paw edema and 12-0-tetradecanoylphorbol-3-acetate-induced ear edema as well as in vitro inhibitory effect on phospholipase-~ enzyme. In some similar studies, various kaempferol glycosides have also shown to possess significant inhibitory activities on TNF -{X, production and nitric oxide (NOS) release (Liang et al., 1999; Authore et al., 2001; Matsuda et al., 2001).
GENTIANACEAE Isoorientin is a common C-glycosyl flavone, luteolin-6-C glucoside, and has been reported in many different plant species such as Gentiana, Asphodelus, Rumex, Swertia and Vitex species (Harborne, 1994). During the studies on a Turkish folk medicine, Gentiana olivieri Griseb. (Gentianaceae), isoorientin was isolated as the active anti-hepatotoxic component from the flowering herbs through in vivo bioassay-guided fractionation procedures against carbon tetrachloride-induced hepatic damage (Deliorman-Orhan et al., 2003). In a following bioassay-guided activity assessment study on the same plant material same compound was again isolated as the active hypoglycaemic component (Sezik et al., 2004). In a reference survey, several other biological activities were attributed to isoorientin including potent antioxidant (Ko et al., 1998), antimicrobial (Afifi et al., 1997), myolitic activity on smooth musclecontaining preparations from the rat and guinea pig uterus (Afifi et al., 1999), etc. Isoorientin (Fig 3) was also shown to possess significant
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
80
antinociceptive and anti-inflammatory activities without inducing any apparent acute toxicity as well as gastric damage. Isoorientin induced inhibition between 28.4-37.1 % and 36.4-43.4% against carrageenan-induced inflammation at 15 and 30 mg/kg doses, respectively. The most remarkable point was the anti-inflammatory activity of isoorientin was found almost equal to that of indomethacin (10 mg/kg) which exerted 30.4-40.3% activity, in a very close dose level. Maybe more important point was isoorientin did not induce any visible gastric damage. Due to the high gastric lesion risk of nonsteroidal anti-inflammatory agents, i.e. indomethacin and acetyl salicylic acid, this is a very important peculiarity. On the other hand, isoorientin was also found to possess significant antinociceptive activity, though not potent as acetyl salicylic acid (Kiipeli et al., 2004). Literature survey demonstrated that isoorientin possesses a wide range of biological activities. Due to the adjacent hydroxyl groups at ring B, i.e. catechol functions, the most prominent biological activity reported for isoorientin is a potent antioxidant activity (Diena et al., 2003). Isoorientin showed a strong antiperoxidative activity towards linoleic acid (Mun'im et al., 2003). The antioxidant activity ofisoorientin was also reported by Budzianowski et al. (1991) through evaluating the inhibitory effect on MDA concentration stimulated by Fe+3 ions and ascorbic acid in rat liver microsomes. Ko et al. (1998) studied the antioxidant activity ofisoorientin-6"-0-glucoside, a watersoluble form, from Gentiana arisanensis (Gentianaceae) using various models and reported a potent antioxidant and radical scavenging activity and suggested as an antioxidant therapy against a wide range of free radicalinduced disorders particularly to protect human LDL from oxidative attack. Isoorientin was isolated as one of the active anxiolytic ingredients from a Peruvian folk remedy, Jatropha cilliata M. Arg. (Euphorbiaceae) at 40 and 100 mg/kg doses and the activity at the later dose was identical to that of diazepam. Under the same conditions, a similar effect was observed for orientin obtained from another fraction, however luteoline, aglycone of these two flavonoids, was ineffective. In the same work, analgesic effect was also investigated and isoorientin was found to have more potent effect than orientin (Okuyama et al., 1996). H
OH
H OH
H
H
OH
Fig 3. Structure ofisoorientin
H
H
81
Effects of Medicinal Plants in Turkey
GERANIACEAE Geranium species are used as antiasthmatic, antiallergic, antioxidant, antidiarrhoeic, antihepatotoxic, diuretic, tonic, haemostatic, stomachic and antidiabetic in folk medicine (Baytop, 1999). G. pratense ssp. finitimum is particularly used to treat stomachache in in Turkey (Ozaydin et al., 2006). As a relevant activity, G. maculatum has been reported to treat duodenal ulcers as well as in diarrhoea and hemorrhoids in Canada (British Herbal Pharmacopeia, 1987). The leaves of G. thunbergii have been used as an antidiarrhoeic in Japan (Fujiki et al., 2003) and G. sanguineum was reported to possess a potent effect against influenza virus (Sokmen et al., 2005). To obtain an experimental evidence on the therapeutic efficacy of Geranium species and its phenolic compounds for inflammatory diseases, the aqueous extract of aerial parts of G. pratense subsp. finitimum (Woronow) Knuth, its fractions and isolated compounds, i.e. the mixture of quercetin 3-0-aR,
OH
HO
OH
1
R}
R2
OH
H H H H H H H OH
H
2
OH OH
3 5
H
6
OH OH OH
0
R3 a-arabinopyranose f3-galactopyranose f3-glucopyranose f3-galactopyranose f3-glucopyranose (2"-O-galloyl)-f3-glucopyranose (2"-O-galloyl)-f3-galactopyranose (2"-O-galloyl)-f3-glucopyranose OH
~
CI
#
OH
'/11111
0 0H
OH
4 (-)-6-chloroepicatechin
Fig 4. The chemical structures ofthe isolated compounds from Geranium pratense subsp. finitimum
82
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
arabinopyranoside, kaempferoI3-0-[3-galactopyranoside (1), the mixture of quercetin 3-0-j3-glucopyranoside, quercetin 3-0-j3-galactopyranoside (2), kaempferoI3-0-j3-glucopyranoside (3), (-)-6-chloroepicatechin (4), the mixture of quercetin 3-0-(2-0-galloyl)-j3-glucopyranoside, quercetin 3-0-(2-0-galloyl)[3-galactopyranoside (5) and myricetin 3-0-(2-0-galloyl)-j3-glucopyranoside (6) (Fig 4) were examined on carrageenan-, PGE 2 - and TPA-induced inflammation in mice and p-benzoquinone induced writhing reflex to assess anti-inflammatory and antinociceptive activities. The effective dose of materials for the inhibition of carragenan-induced hind paw edema assay was determined to be 100 mg/kg, which was also used in the assays with the extract, its fractions and isolated compounds in all other experiments. The aqueous extract as well as compounds 1 and 2 (100 mg/kg) significantly inhibited the formation of the carrageenan-induced hind paw edema. Significant reductions were also exerted by the aqueous extract and the compounds 1,2 and 5 against PGE 2-induced hind paw edema and TPAinduced ear edema models. The same compounds were shown to possess antinociceptive activity (Kupeli et al., 2007 d). JUGLANDACEAE
Juglans regia L. leaves have been used mostly in worldwide traditional medicines as antifungal, antihelmintic, astringent, keratolytic, antidiarrhoeal, hypoglycaemic, depurative, tonic and for the treatment of sinusitis, cold and stomachache (Girzu et al., 1998; Mouhajir et al., 2001). In Turkish folk medicine, however, leaves are frequently used to reduce fever in sunstroke or to alleviate rheumatic pain, externally. Fresh leaves are spread to form a bed for unclothed patient, who is covered with fresh leaves or pounded with/without raw salt and applied on the naked body or forehead to reduce fever or on swelled joint to alleviate the rheumatic pain, or boiled in water and used for bathing in both conditions (Fujita et al., 1995; Ye~ilada, 2002). Both aqueous and EtOH extracts showed significant antinociceptive activity without inducing any gastric damage, but the effect of later extract was more pronounced than the former as well as that of aspirin (Erdemoglu et al., 2003). EtOH extract also showed a potent antiinflammatory activity as potent as indomethacin. Since direct application of fresh leaf is suggested for the treatment, less polar components of the leaves might possibly be more active (Erdemoglu et al., 2003). LAMIACEAE Prunella vulgaris L. is used against rheumatism, colds and cardiac disorders in Turkish folk medicine (Yesilada et al., 1993). In a previous study, a herbal combination (SKI 306X) consisting of Clematis mandshurica, Trichosanthes kirilowii and Prunella vulgaris, which was used in Chinese traditional medicine for the treatment of various inflammatory disorders, i.e. lymphadenitis and arthritis, was studied for their in vitro effects as anti-inflammatory, analgesic,
Effects of Medicinal Plants in Turkey
83
antiarthritic, blood microcirculation-enhancer as well as inhibition of cartilage degeneration enzyme activities on proteoglycan degradation in cartilage explant culture and collagenase-induced rabbit ostheoarthritis model (Choi et al., 2002). The results revealed that SKI 306X inhibited proteoglycan degradation in a concentration-dependent manner and it also showed protective effect on the knee joint of rabbit from ostheoarthritis-like changes on prophylactic administration. In another study, the aqueous extract of P. vulgaris was shown to possess strong antioxidant potency in inhibiting rat erythrocyte hemolysis and lipid peroxidation in rat kidney and brain homogenates (Liu & Ng, 2000). On the other hand, in a study was performed by our group, in vivo anti-inflammatory or antinociceptive activity of the plant was found to be weak (Kupeli et al., 2007). The genus Lamium L. (Lamiaceae) is characterized by almost 40 annual or perennial herbaceous plants spread throughout Europe, Asia, and Africa (Willis, 1973). Lamium album, L. maculatum, and L. purpureum exhibit antispasmodic, anti-inflammatory, antioxidant and antiproliferative properties and have been extensively used in folk medicines as a remedy for the treatment of several disorders such as trauma, fractures, putrescence, paralysis, leucorrhoea, hypertension, as well as women affiictions, such as menorrhagia, uterine hemorrhage, vaginal or cervical inflammations (Bremness, 1995; Matkowski & Piotrowska, 2006; Paduch et al., 2006; Shuya et al., 2003; Weiss, 1988). In Turkish flora, thirty Lamium species have been recorded (Duman, 2000a; Mill, 1982). Among them, L. album, L. maculatum, and L. purpureum have been reported to be used as tonics and for the treatment of constipation in Anatolia (Baytop, 1999). In western Anatolia (in particular in Manisa) whole plant of L. album and several other Lamium species are used to relieve pain in rheumatism and other arthritic ailments (Ozaydin et al., 2006). In order to assess the traditional usage various extracts were prepared from several Lamium species. The n-butanol extracts of L. garganicum subsp.laevigatum (LGL-BuOH), L. garganicum subsp. pulchrum (LGP-BuOH), and L. purpureum var. purpureum (LPPBuOH) exhibited notable inhibition (16.5-28.9%, 14.5-26.9%, 12.3-21.5%, resp.) in carrageenan-induced hind paw edema model at doses of200 mg/kg without inducing any gastric damage. The LGL-BuOH (7.1-30.4%) and LGPBuOH (5.9-24.1%) extracts also displayed potent anti-inflammatory activity against PGE 2-induced hind paw edema model. However, the remaining extracts were found to be inactive against carrageenan-induced hind paw edema and PGE 2-induced hind paw edema models. On the other hand, all the extracts of the title plants failed to display any anti-inflammatory activity on TPA-induced mouse ear edema model. LGL-BuOH and LGP-BuOH were also found to exhibit remarkable antinociceptive activity inp-benzoquinoneinduced abdominal constriction test at a dose of200 mg/kg (25.0% and 24.3%, respectively). The experimental data demonstrated that L. garganicum subsp. laevigatum and L. garganicum subsp. pulchrum displayed remarkable anti-inflammatory and antinociceptive activities (Kiipeli Akkol et al., 2008b).
84
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Salvia species, Sage, features prominently in the national pharmacopoeias of many countries throughout the world and several species have been used as a remedy against perspiration and fevers; as a carminative; a spasmolytic; an antisepticlbactericidal; an astringent; as a gargle or mouthwash against the oral inflammations such as tongue or throat; a wound-healing agent; in skin and hair care; and against rheumatism (Kintizios, 2000). S. virgata is known as "yilancik" in Turkey and used to alleviate dermatological problems and wounds. The decoction of S. virgata is used against blood cancers in Western Turkey (Baytop, 1999). S. halophila is an endemic plant of Turkey and traditionally being used as herbal tea. To provide a scientific evidence for traditional uses, the aerial parts of S. halophila and S. virgata were subjected to Soxhlet extraction with different solvents i.e. n-hexane, ethyl acetate, methanol and aqueous methanol (50%). Plants were also extracted with water under reflux. The effects ofthe extracts were studied inp-benzoquinone-induced abdominal constriction test for the assessment of antinociceptive activity and carrageenan-induced hind paw oedema and 12-0-tetradecanoyl-13acetate (TPA)-induced ear oedema models in mice for assessing the antiinflammatory activity. Results showed that methanol extract significantly inhibited carrageenan-induced paw oedema and p-benzoquinone-induced abdominal constrictions at 100 mglkg dose, while it was ineffective against the TPA-induced ear oedema. However, the other extracts did not show any inhibitory antinociceptive and anti-inflammatory activities on these in vivo models. The extracts were then analysed using a HPLC-PDA method. Rosmarinic acid was found as the main constituent in the extracts, while caffeic acid and luteolin derivatives were also detected in small ratio (Kiipeli Akkol et al., 2008a). In a literature survey, Hosseinzadeh et al. (2003) reported that the aqueous extract of Salvia leriifolia seeds showed significant and dose-dependent (1.25-10 g/kg) antinociceptive activity over 7 h, and the activity was inhibited by naloxone pretreatment in hot-plate and tail flick tests. They also observed significant and dose-dependent (2.510 glkg) activity against acute inflammation induced by acetic acid and in the xylene ear oedema test. In the chronic inflammation test (cotton pellet test), however, the extract (2.5-5 g/kg) demonstrated considerably significant and dose-dependent anti-inflammatory activity. Consequently, a supraspinal antinociceptive effect mediated by opioid receptors has been suggested as the activity mechanism of extract (Hosseinzadeh et al., 2003). On the other hand, in a previous study, the methanol extract of Salvia miltiorrhiza was found to inhibit production ofPGD 2 and the ethyl acetate subfraction exerted the strongest inhibition. From the ethyl acetate subfraction, tanshinone I was isolated as an active principle through activity-guided isolation procedures (Kim et al., 2002). The genus Sideritis L., widely distributed in MediterraneanMacronesian region, are traditionally used in Spanish folk medicine for their anti-inflammatory and gastroprotective properties to treat certain
Effects of Medicinal Plants in Turkey
85
disorders that are accompained by inflammation. Several anti-inflammatory compounds have been obtained from the members of this genus, mainly flavonoids and terpenoids (Godoy et al., 2000). In Turkey, this genus is represented by 46 species (Duman, 2000b; Ayta!; & Aksoy, 2000) and some of which are used in the treatment of gastrointestinal ailments, common colds (including bronchitis, sore throat, flu) and as a diuretic as well as a herbal tea for pleasure in Turkish folk medicine (Baytop, 1999; Yesilada et al., 1995). In the light oftraditonal usage, acetone extract from the aerial parts of Sideritis ozturkii Aytac & Aksoy and its fractions were investigated for its in vivo anti-inflammatory and antinociceptive activities. Acetone extract of the plant and its phenolic fraction were found to possess significant inhibitory activity on carrageenan-induced hind paw oedema and pbenzoquinone induced writhing reflex models in mice. Ozturkoside A (chrysoeriol 7-0- [2'"-0-caffeoyl-6'"-O-acetyl-13-D-glucopyranosyl-( 1~2)-13- Dglucopyranoside]) (1), ozturkoside B (chrysoeriol 7-0-[2"'-0-caffeoyl-13-Dglucopyranosyl-(1~2)-13-D-glucopyranoside]) (2) and ozturkoside C (chrysoeriol 7-0- [2'"-0-p-coumaroyl-6'"-O-acetyl-13-D-glucopyranosyl-( 1~2)13-D-glucopyranoside]) (3) were isolated from the active phenolic fraction (Fig 5). Ozturkoside C showed notable antinociceptive and anti-inflammatory activities without inducing any apparent acute toxicity or gastric damage.
OH
OH
HO
0
a
Fig 5. Chemical structures of compounds 1-3
Although the activity of ozturkosides A and B were not significant in statistical analysis, some inhibitory effect was observed. Accordingly, it is suggested that these components in phenolic fraction might possibly share the antinociceptive and anti-inflammatory activities together (Kiipeli et al., 2007b). In spite of a high number of studies reporting the anti-inflammatory and antinociceptive activities of several Sideritis species only two flavonoids have been isolated and defined as the active constituents; hypolaetin-8-
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
86
glucoside (Villar et al., 1984) and 5-0-demethylnobiletin (Bas et al., 2006). As shown in Fig 6, hypolaetin-8-glucoside shows very close chemical structure to ozturkoside C, both having luteoline type flavone glycoside structure. OH OH O-Glu HO
o OH
Fig 6. Chemical structure ofhypolaetin-8-glucoside
Additionally, acetone extract of S. stricta Boiss. & Heldr. apud Bentham and its phenolic fraction exhibited potent inhibitory activity against both bioassay models in mice. From the active phenolic fraction a well-known phenylethanoid glycoside, verb as cos ide (= acteoside) (1) and two flavonoid glycosides, isoscutellarein 7-0- [6'" -O-acetyl-~- D-allopyranosyl(1 ~2)l-~-D-glucopyranoside (2), and isoscutellarein 7-0-[6'" -O-acetyl-~-D allopyranosyl-( 1~2)l-6f1-0-acetyl-~- D-glucopyranoside (3) were isolated. During phytochemical studies we also isolated a methoxyflavone, xanthomicrol (4) from the non-polar fraction (Fig 7). Although antinociceptive and anti-inflammatory activities of phenolic components were found not significant in statistical analysis, compounds 1 to 3 showed a notable activity without inducing any apparent acute toxicity as well as gastric damage. Furthermore, a mixture of flavonoid glycosides (2 + 3) exhibited a significant inhibitory effect in both models at a higher dose (Kiipeli et al., 2007c).
HO
HO
1
87
Effects of Medicinal Plants in Turkey Fig 7. Contd. R OH HO
OCOCH3
o o
HO OH
2 R:OH 3 R: OCOCH 3 4'
OH
OH
0 4
Fig 7. Chemical structures of compounds 1 - 4
LAURACEAE Laurus nobilis L., laurel, is a spice plant widely distributed in the Mediterranean area and the seed oil is used against rheumatic pain in Turkish folk medicine as well as in Europe. Previously, the oil obtained from laurel fruit was also reported to be used for treatment of furuncles, sprains, bruises and rheumatism (Simic et al., 2003). The experimental results indicated that the ethanol extract of the seeds possesses a remarkable anti-inflammatory (29.1-37.3%) and antinociceptive (34%) activity at a dose of 500 mg/kg (Kupeli et al., 2007c). On the other hand, extract prepared from the leaves were also studied for comparison and was found completely devoid of any activity (Kupeli et al., 2007 c).
LORANTHACEAE The genus Viscum L. (Loranthaceae) comprises semi-parasitic plants which grow on various host tree and shrubs (Miller, 1982). Viscum album L. is the most widespread species worldwide and has been reputed against cardiovascular diseases, i.e. hypertension and atherosclerosis; various bone and joint disorders including periarthritis, spondylitis, and arthritis; to
88
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
alleviate headache; for immune system stimulation; in nervous disorders as sedative or to combat epilepsy (Bartram, 1995; Murray, 1995; Wichtl & Bisset, 1994). In order to evaluate this information, antinociceptive and anti-inflammatory activity ofthe five flavonoids [5,7 -dimethoxy naringenin (1,3, and 5) or 4', 6'-dimethoxy chalcononaringenin (2, 4)] derivatives; (5,7dimethoxy-flavanone-4' -0- ~- D-gi ucopyranoside (1), 2'-hydroxy-4' ,6'dimethoxy-chalcone-4-0-~- D-glucopyranoside (2), 5,7 -dimethoxy-flavanone4'-0- [2"-0-(5111-0-trans-cinnamoyl)-~-D-apiofuranosyl]-~- D-glucopyranoside (3), 2'-hydroxy-4' ,6'-dimethoxy-chalcone-4-0-[2"-O-(5'"-O-trans-cinnamoyl)-~ D-apiofuranosyl]-~- D-glucopyranoside (4), 5,7 -dimethoxy-flavanone-4'-0- [~ D-apiofuranosyl(I~2)]-~-D-glucopyranoside (5) isolated from ethyl acetate fraction of V. album ssp. album were investigated (Fig 8). The ethyl acetate fraction showed a remarkable and dose-dependent anti-inflammatory activity on carrageenan-induced hind paw oedema model (between 24.7 to 37.2%). Same pharmacological profile was also observed for the compound 2 (30.7-33.6%) from the ethyl acetate fraction. The anti-inflammatory activity of 2 was almost as potent as that of indomethacin which exerts 33.3-42.7% anti-inflammatory activity. Other flavonoids, 1, 3, and 5, isolated from the ethyl acetate fraction, also showed significant but weaker antiinflammatory activity against the same model. Among the isolated compounds from the ethyl acetate fraction were administered in 30 mg! kg dose, compounds 2 and 5 were found to possess highest and significant antinociceptive activity on p-benzoquinone induced writhing reflex test, but not as potent as ASA. The ethyl acetate fraction and isolated compounds did not induce any apparent acute toxicity during the 24 h observation period. It is noteworthy that, in spite of a weak gastric lesion incidence in the higher dose of ethyl acetate fraction (gastric lesions were observed in the stomach of lout of 6 rats), all the isolated flavonoids were found completely safe from the view point of gastric damage (Deliorman et al., 2006). Herencia et al. (1999), investigated the effects of a series of chalcone derivatives on various in vivo and in vitro inflammatory models (i.e. human neutrophil functions, eicosanoid release, TNF -ex production, air-pouch, etc.) and reported inhibitory effects on iNOS and COX-2. Correa et al. (2001) reported potent antinociceptive activity for 3,4-dichlorochalcones using the writhing test in mice. In the study of Pelzer et al. (1998) it was concluded that the anti-inflammatory activity offlavonoids increase depending upon the catechol or guaiacol-like substitution to Bring (3', 4'-dihydroxy or 3"hydroxy-4'-methoxy or 3'-methoxy-4'-hydroxy). Accordingly flavanones such as eriodictyol, 7-0-methyleridictyol and hesperidin showed the highest activity against carrageenan-induced edema. However, flavanone and chalcone derivatives demonstrated to possess anti-inflammatory activity in the present study were shown to possess one glucosylated hydroxyl group at Bring [5, 7-dimethoxy naringenin (1, 3, and 5) or 4', 6'-dimethoxy chalcononaringenin (2, 4) derivatives]. Therefore, results showed herein present additional structural data for the anti-inflammatory
Effects of Medicinal Plants in Turkey
89
and antinociceptive activity evaluation of chalcone and flavanone derivatives.
8
°
~9
6
5
~o
4
° (1)
HO HO~OH
14°"f-°----i-CH20H OH
5
°
(2)
°
°
or~/~f HO HO
(4)
Fig 8. Contd.
90
Camp. Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
Fig 8. Contd.
HO~'''' ~"
I
1 """"-
2
~'HO OH
3' 4'
6
0
1" 0
CH20H
5
3
(5)
Fig 8. Structures of investigated compounds
Arceuthobium oxycedri (DC.) Bieb., is a semi-parasitic plant grows on Juniperus oxycedri and the shape and color ofthe plant looks very similar to host plant. In Anatolia this plant is believed to be effective against a wide range of diseases; from inflammatory to infectious (Yesilada et al., 1995; Fujita et al., 1995; Honda et al., 1996). To confirm the conventional usage, prepared ethanol and aqueous extracts of the plant were studied and both extracts exhibited significant anti-inflammatory effect at the ranges of 28.9-34.2% and 21.1-30.5%, respectively. The plant also showed a potent antinociceptive activity; for the aqueous extract 34.2% at 500 mgt kg dose, while for the ethanol extract 28.6% at 250 mg/kg and 39.9% at 500 mg/kg doses. Results supported the folkloric utilization of the plant as no such study has been carried out on this plant previously (Kupeli et al., 2007c).
LYCOPODIACEAE Lycopodium genus (syn. Huperzia), commonly known as "club moss, ground pine, devil's claw, devil ash" in English, is a pteridophyte abundantly found in subtropical and tropical forests in the world, which is under the risk of extinction (Lawrence, 1989). In Turkey, the genus is represented by five species, namely L. alpinum L., L. annotinum L., L. clavatum L., L. complanatum ssp. chamaecyparissus (A. Br.) Doll, andL. selago L. (Davis & Cullen, 1984). Among them, L. clavatum (LC), and has been reported to be used for wound-healing effect as well as against nappies occurring in babies and, therefore, also called "belly powder" (Baytop, 1999). The spores ofthe plant were stated to have a protective effect as dusting powder for tender skin (Vasudeva, 1999). In Papua New Guinea, LC is used against stomach pain (Holdsworth & Giheno, 1975). Some other Lycopodium species have also been reported for similar or related ethnobotanical utilizations
Effects of Medicinal Plants in Turkey
91
worldwide. According to an ethnobotanical survey conducted in Sabah region of Malaysia, Lycopodium sp. was recorded to be used to treat skin burns (Kulip, 1997). In Wisconsin (USA), the leaf decoction of L. obscurum was reported to be used against muscle pain and rheumatism (Coffey, 1993). Studies on four extracts prepared with petroleum ether, chloroform, ethyl acetate and methanol as well as the alkaloid fraction from the aerial parts of L. clavatum L. using acetic acid-induced increase in capillary permeability assessment in mice revealed that only the chloroform extract and the alkaloid fraction displayed marked anti-inflammatory effect at a dose of 500 mg/kg having percentage of inhibition 24.3 and 32.1, respectively, as compared to indomethacin which exhibited 44.6% of inhibition at 10 mg/kg dose. The alkaloid fraction of L. clavatum was submitted to bioassay-guided fractionation procedures and revealed that the alkaloidal-type of compounds might possibly be responsible for the anti-inflammatory activity, which supports the folk medicinal utilization of the plant. Gas chromatographicmass spectrophotometric analysis of the active alkaloid fraction have demonstrated that lycopodine, an alkaloid isolated from several Lycopodium species, was the major alkaloid component (84.5%) (Fig 9). Accordingly, these experimental results point out that the anti-inflammatory activity of the alkaloid fraction of the aerial parts of Lycopodium clavatum are primarily due to the alkaloidal components, which might most probably be lycopodine as the major responsible compound (Orhan et ai., 2007a).
Fig 9. Chemical structure oflycopodine
LYTHRACEAE
Lythrum saiicaria (purple loosestrife) was known as medicinal plant already in ancient Greek and Roman times and it has been an important drug for centuries. The whole flowering plant and the flowering branch tips of this
92
Camp. Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
plant are used not only in folk medicine but also in pharmaceuticals. Decoction or some other fluid extracts from Lythrum salicaria is used internally for diarrhoea, chronic intestinal catarrh, hemorrhoid and eczema. It is also used externally to treat varicose veins, bleeding of the gums, hemorrhoids and eczema (Baytop, 1999; Mantle et al., 2000; Rauha et al., 2000). Dried aerial parts of L. salicaria L. were sequentially extracted with different solvents such as petroleum ether, ethyl acetate, methanol and 50 % aqueous methanol. Water extract of L. salicaria was also prepared under reflux. The extracts were then submitted to bioassays, i.e. free radical scavenging activity [1,1-diphenyl-2-picrylhydrazyl, DPPH% assay, iron(IlI) reductive activity, capacity of the inhibition of linoleic acid peroxidation and MDA formation], antinociceptive activity fp-benzoquinone-induced abdominal constriction test] and anti-inflammatory activity [carrageenaninduced hind paw edema model]. In addition, the content oftotal phenolics, flavonoids and flavonols in the extracts were determined by spectrophotometric methods. Although the methanol extract showed antiinflammatory and antinociceptive activity other extracts remained inactive. None of the extracts caused any gastric damage. The qualitative and quantitative compositions of all the extracts were analysed using a HPLCPDA system, polar fractions were found to be rich in flavonoids such as isovitexin and isoorientin (Tunalier et al., 2007). In a literature review, L. salicaria tannins, flavone-C-glycosides and anthocyanins have been identified in L. salicaria extracts. Isoorientin, isovitexin and their derivatives have been reported as its main flavonoids (Rauha et al., 2000; Rauha, 2001). Rauha (2001) and Tunalier et al. (2007) reported the concentration of flavonoids in L. salicaria to be high and isoorientin was reported to possess anti-inflammatory while isovitexin antioxidant activities (Ko et al., 1998; Arimoto et al., 2000; Kiipeli et al., 2004). MORACEAE
Maclura pomifera (Rafin.) Schneider (syn. M. aurantiaca Nutt.), Ioxylon pomiferum Raf., Toxylon pomiferum Raf. ex Sargis a widely cultivated hardwood tree in Turkey for ornamental purposes. Various parts of the Maclura species are used in folkloric medicine worldwide. Decoction prepared from the roots of M. pomifera is used for the treatment of sore eyes by Comanche Indians in the North America (Carlson & Volney, 1940). The sap ofthe plant is used for the treatment of toothache and the barks and leaves for uterine haemorrhage in Bolivia (Bourdy et al., 2004). While the bark of M. tinctoria has been reported to be used against toothache by Kaiowa and Guarani indigenous people living in the Caarapo Reserve in Brazil as well as the in the other parts of Amazon region, it was also recorded to be used
Effects of Medicinal Plants in Turkey
93
in southern Ghana for dental health (Elwin-Lewis et al., 1980; Elvin-Lewis & Lewis, 1983; Bueno et al., 2005). Aqueous, ethanolic and chloroform extracts and two prenylated isoflavones; scandenone CD and auriculasin (II), isolated from the fruits of Maclura pomifera, were investigated for their in vivo anti-inflammatory and antinociceptive activities. The ethanol and chloroform extracts and scandenone had 37.2%, 42.0% and 29.7% inhibition, respectively, while acetylsalisilic acid (ASA), the reference compound, showed 53.5% inhibition at 100 mg/kg dose. Extracts and scandenone also exhibited significant inhibition, ranging between 23.3-41. 7% for the ethanol and 16.940.3% for the chloroform extracts as well as 23.9-35.4% for scandenone in carrageenan-induced hind paw edema model at 100 mg/kg dose. All the extracts and compounds were found completely safe from the viewpoint of gastric damage and no acute toxicity was observed within 48 h observational period. A similar activity pattern was also observed in TPA-induced ear edema model, the EtOH extract (49.2% and 54.9%), chloroform extract (45.1 % and 49.6%) and scandenone (45.7% and 49.3%) displayed potent antiinflammatory activity as measurement of edema weight as well as swelling thickness, compared to indomethacin (62.7% and 70.5%). However, the H 20 extract and auriculasine were found to be completely inactive against this model. Results of the present study have revealed that isoflavonoids are the major anti-inflammatory and antinociceptive principles of the fruits. Scandenone showed a potent activity profile against all the models employed, while that of other isoflavonoid, auriculasin, was not significant, but within reasonable limits, i.e. 20.6% on p-benzoquinone-induced writhing test (Kiipeli et al., 2006). Among the various biological activity profiles reported for flavonoids so far, potent antioxidant activity was particularly attributed to the existence of a catechol group in the B ring (Brandi, 1997; Hom-Ross et al., 2003; Tsao et al., 2003; Cioffi et al., 2003). Since inflammation has been found to elevate the levels offree radicals, antioxidant agents might be also considered possibly to display anti-inflammatory effect (Guo et al., 2002). As shown in Fig 10, auriculasin possesses a catechol group in the molecule; however the anti-inflammatory activity was less potent, not significant, than that of scandenone which does not possess this function. Tsao et al. (2003) studied the antioxidant activity ofisoflavones from M. pomifera fruits. Accordingly, they isolated two linear isoflavones; osajin and pomiferin, as the main constituents from the ethyl acetate extract. These compounds were chemically constitutional isomers of scandenone and auriculasin. Pomiferin, possessing catechol group on ring B, was found to display a strong antioxidant activity, as expected, comparable to the antioxidant vitamins C and E, while osajin (bearing single hydroxyl group in ring B), genistein and daidzein showed no antioxidant activity.
94
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
o R
OH
o
R=H (scandenon) R=OH (auriculasin)
OH
Fig 10. Chemical structures of scandenone and auriculasin
POLYGONACEAE Rumex crispus L. is reported to treat headache, wound healing and to promote maturation of abscess in folkloric medicine in Turkey (Yesilada et al., 1995). Besides, a literature survey revealed that several other Rumex species have also been traditionally used in the treatment of eczema, wound healing, swelling, tonsillitis, leprosy and hemorrhoids in Ethiopia or as analgesic and antipyretic in India and to treat burns and to remove pain from the back and lumber region in Bangladesh (Gebrie et al., 2004; Ghosh et al., 2003; Rouf et al., 2003). In a previous study on the anti-inflammatory activity oftwo Rumex species based on the cell viability and measurement ofPGE 2 concentration, the results verified that the root extract of R. abyssinicus to inhibit PGE 2 synthesis, whereas the leaf extract of R. nervosus showed no inhibition at all (Getie et al., 2003). In another study, anti-inflammatory activity of R. patientia, a plant used for wound healing and anti-inflammatory properties in Turkish folk medicine, was studied against carrageenan, histamine, dextrane, serotonin, formaldehyde-induced oedema, cotton pellet granuloma and Kabak test models in rats and the extract was found to possess significant anti-inflammatory activity which was concluded to be dependant on its rich anthraquinone and tannin content (Suleyman et al., 1999). Conversely, the experiments performed by our group, demonstrated that both aqueous and ethanol extracts of R. crispus displayed no remarkable anti-inflammatory or antinociceptive activity (Kupeli et al., 2007). RANUNCULACEAE The Clematis species have been extensively used in the traditional medicines worldwide due to its widespread distribution at the northern hemisphere. The aerial parts of various Clematis species are particularly used in Europe
Effects of Medicinal Plants in Turkey
95
and Eastern Asia as a remedy to reduce pain and fever, as diuretic, used in the treatment of rheumatic pain, eye infections, gonorrhoeal symptoms, bone illnesses, chronic skin disorders, gout and varicosity (Keys, 1985; PDR, 2000). Several species of Clematis have been used as remedy in Anatolia as well. The most salient application related to folkloric usage is the one that is applied in the treatment of rheumatic ailments. In Northern Anatolia, C. cirrhosa vs C. flammula leaves or the aerial part are used to provide temporary relief of joint pains. Ground fresh aerial parts or leaves are applied on inflammatory joints for about 15-30 minutes. The ensuing irritation on the skin opens a gap and drains the edema. In some particular cases, the wound is plugged by inserting a grape dregs in order to provide continuous drainage of the edema for 20-25 days. The plug is removed occasionally to drain the accumulated inflammation out. Then, to cure this open wound a fresh leaf of Plantago major ssp. major L. is applied (Yesilada et al., 1991). Moreover, C. vitalba branches are also used to stop tooth pain by smoking like a cigarette in northwestern Anatolia (Yesilada et al., 1999). In order to test the effectiveness of extracts, fractions and subfractions from dried Clematis vitalba L. aerial parts were studied in mice. Ethanolic extract exerted a potent effect on carrageenan-induced hind paw edema and acetic acid-induced increased vascular permeability models. Through bioassay-guided fractionation procedures a new C-glycosyl flavone, 4'-0coumaroyl-isovitexine (vitalboside) was isolated as the main active ingredient ofthe aerial parts (Fig 11). Vitalboside showed a potent and dose-dependent (in 75 and 150 mg/kg does, per os) in vivo anti-inflammatory activity against acute (carrageenan-, serotonin- and PGE 2-induced hind paw edema model, castor oil-induced diarrhoea), subacute (subcutaneous air-pouch) and chronic (Freund's complete adjuvant-induced arthritis) models of inflammation. The same compound was also isolated as the main antinociceptive principle which was assessed by using the models based on the inhibition ofp-benzoquinoneinduced writhings, as well as antipyretic activity against Freund's complete adjuvant-induced increased body temperature. Acute and subchronic toxicity studies were also performed (Yesilada & Kiipeli, 2007). Previously, Li et al. (2002,2003) studied the in vitro activity of several plants on cyclooxygenase1 (COX-l) enzyme, one of the key enzymes in inflammatory pathway, including Clematis species which are used against various inflammatory disorders such as asthma, arthritis, rheumatism, fever or snake bite in Australia and China. They reported that C. chinensis and C. glycinoides stems did not show any apparent activity against COX-1 enzyme, while C. pickeringii leaves were found to possess a potent inhibition. In spite of the worldwide practice of using dried aerial part of Clematis species against inflammatory disorders in traditional medicines, fresh aerial part of Clematis species is known to possess severe irritating in nature. Such irritation caused by the enzymatic transformation of the ranunculin glycoside into protoanemonin after crushing the fresh plant (PDR, 2000). Ranunculin and eventually protoanemonin which are also known as the most characteristic
96
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
components of some other Ranunculaceae plants such as Ranunculus, Anemone or Helleborus species are ultimately irritating and volatile substances (Didry et al., 1993; Southwell & Tucker, 1993). In fact, some Ranunculus species are also practiced in the same way as of Clematis species for the same purpose among locals in Anatolia (Yesilada et al., 1995). Protoanemonin is an utterly weak substance and transforms into anemonin after being rapidly dimerized. On the other hand, no protoanemonin has been found in the dried plant (PDR, 2000). At the monograph of the plant (Traveller's Joy) in PDR Gruenwald et al., (2000) for Herbal Medicines it is reported that no side effect was observed on either internal or external application of the dehydrated plant, while even contact with the crushed or cut fresh aerial part induces irritation on the skin. Accidental consumption ofthe fresh aerial part leads to strong irritation at the gastrointestinal and urinary tract and provokes colic, diarrhoea and related symptoms. Higher doses are reported to induce death on experimental animals depending upon the protoanemonin content in the plant. On the other hand, Pieroni et al. (2002) reported that C. vitalba sprouts are boiled in large amounts of water in order to cleave protoanemonin prior to eaten in central Italy.
OH
OH Fig 11. Chemical structure of 4' -O-coumaroyl-isovitexine <=vitalboside)
In Turkish folk medicine, Helleborus orientalis Lam. roots are used to treat edema in cattle. A small piece of root is sharpened like a nail is inserted into a hole punched in the ear/or tail of cattle and kept 24 h to reduce inflammation in extremities or applied externally on swelled joints or dried and pounded root is orally administered (Fujita et al., 1995; Yesilada, 2002). Alternatively, the extract of roots mixed with Salvia tomentosa herbs is used as a bath for both man and animals. Although roots ofthe plant are preferentially used to alleviate the inflammatory conditions in cattle, it is not restricted to animals. A small piece of root is inserted inside a cavity against toothache in man (Fujita et al., 1995). The utilization of another Helleborus species, H. purpurascens, for the treatment of inflammatory stages of rheumatism and autoimmune diseases has also been reported in Romania (Olinescu et al., 1986). This plant was also shown to have in vivo anti-inflammatory activity in rheumatism as well as immunosuppressant
Effects of Medicinal Plants in Turkey
97
activity (Linke et al., 1998). Another species, H. niger, was also reported to possess immunomodulating properties (Bussing & Schweizer, 1998). In order to assess the anti-inflammatory and antinociceptive activities aqueous and ethanolic extracts were prepared. Our experiments proved that both roots and herbs of H. orientalis to possess significant antinociceptive activity, but that of EtOH extract was more pronounced and did not induce any gastric damage. However, only the EtOH extract of roots showed potent antiinflammatory activity (Erdemoglu et al., 2003). Ranunculus species are used in Turkish folk medicine against rheumatic pain, in particular to drain the pus from the inflammed joint through applying the fresh flowering herb after pounding on joint for about 20 min (Baytop, 1999; Yesilada et al., 1995). In an in vivo study, ethanol extract from R. tricophyllus Chaix herb showed a weak anti-inflammatory and antinociceptive activity (Kupeli et al., 2007).
ROSACEAE The leaves of Laurocerasus offficinalis Roemer are used for asthma, coughs, indigestion and dyspepsia (Grieve, 1994). The fresh leaves of this plant is used in Turkish folk medicine as analgesic for headache and as antipyretic externally. Fresh leaves are applied on the forehead, after being wilted over a fire (Yesilada et al., 1999). Conventional usage of this plant in folk medicine led us to search the biological activity of the plant extracts in a scientific platform. Even though the activity of aqueous extract was weak, EtOH extract showed potent antinociceptive and anti-inflammatory activity without inducing any gastric lesion which supports the claimed activity (Erdemoglu et al., 2003). Different parts of Rubus species are used in folk medicine for the treatment of numerous diseases such as diabetes mellitus, inflammatory disorders, diarrhoea, hemorrhoids and ulcers (Jouad et al., 2002; Marquina et al., 2002; Panizzi et al., 2002). Recently, the novel anti-inflammatory triterpenoids, tormentic acid and euscaphic acid, were isolated from the fresh leaves of R. sieboldii (Murakami et al., 2002). In a previous study, the aerial parts of R. sanctus, R. hirtus and their hybrid were evaluated for anti-inflammatory activity using carrageenan-induced hind paw edema in mice and polar fractions, n-butanol and remaining aqueous fractions obtained by solvent extraction, were shown to possess significant activity (Akcos et al., 1998). Decoction ofthe roots Rubus sanctus is used to alleviate pain and against rheumatism as tea (Honda et al., 1996). In an experimental study, EtOH extracts of R. sanctus Schreber (root and aerial part) and R. hirtus Walds. et Kit (aerial parts) showed potent antinociceptive activity, while that of aqueous extracts were weak. However, it should be noticed that both plants extracts may induce gastric damage. Meanwhile, neither roots nor aerial parts of both species showed a significant anti-inflammatory
98
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
activity against carragenan-induced hind paw edema in 500 mg/kg dose (Erdemoglu et al., 2003). In the German Commission E Monographs, fruits (rose-hips, with seeds) of Rosa canina L. (Rosaceae), the dog rose, are stated to possess prophylactic and therapeutic activities against a wide range of ailments, including the inflammatory disorders arthritis, rheumatism, gout, and sciatica, for diseases with fever; for colds and infectious diseases including influenza, against gastro-intestinal disorders, to aid digestion, prevention of inflammation of the gastric mucosa and gastric ulcer, for gallstones, biliary complaints, as a laxative, for disorders of the kidney and the lower urinary tract, as a diuretic, for dropsy and as an astringent (Blumenthal et al., 1998). The plant has a widespread distribution in Turkey and was reported as the most commonly utilized remedy following Plantago species in Turkish folk medicine (Yesilada, 2002). In addition to the effects offruits against the disorders described above, the fruit is known as the most effective remedy against hemorrhoids and diabetes mellitus in Turkish folk medicine. Besides, the roots and leaves of the plant have also been used against bronchitis (Tabata et al., 1994; Fujita et al., 1995; Yesilada et al., 1995, 1999; Honda et al., 1996; Sezik et al., 2001). This knowledge led us to analyze the plant extracts for their biological activity. The aqueous and ethanol extracts of Rosa canina L. (Rosaceae) fruits and the fractions prepared from the latter were investigated for their anti-inflammatory and antinociceptive activities in several in vivo experimental models. The ethanolic extract was shown to possess significant inhibitory activity against inflammatory models (i.e. carrageenan-induced and PGE1-induced hind paw edema models, as well as on acetic acid-induced increase in a capillary permeability model) and on a pain model based on the inhibition of pbenzoquinone-induced writhing in mice. The active EtOH extract was then fractionated through solvent extractions with increasing polarity and five fractions were obtained; hexane, CHC13 , ethylacetate (EtOAc), n-BuOH, and the remaining water fraction. Ethylacetate and n-butanol fractions displayed potent anti-inflammatory and antinociceptive activities at a dose of 919 mg/kg without inducing acute toxicity. Further attempts to isolate and define the active anti-inflammatory and antinociceptive component(s) from the EtOAc and n-BuOH fractions through bioassay-guided isolation procedures have resulted in failure. Both fractions were further submitted to chromatographic separation, but the activities weakened to a large extent, probably due to the synergistic interaction of components in the active fractions (Deliorman Orhan et al., 2007).
SCROPHULARIACEAE Since the ancient times, Verbascum extracts, decoctions and infusions, commonly known as "mullein", have been used as medicinal herbs in traditional medicines worldwide. The leaves and flowers of Verbascum are
Effects of Medicinal Plants in Turkey
99
reported to have expectorant, mucolytic and demulcent properties which are used to treat respiratory problems such as bronchitis, dry coughs, tuberculosis and asthma in traditional Turkish medicine (Baytop, 1999; Turker & Camper, 2002). Verbascum flowers are boiled in milk and are applied externally for pruritic conditions affecting the urogenital organs. In addition, as a traditional Anatolian cure for anal fistulae, the flowers are boiled in a cauldron and then the anus is exposed to the vapors. These species are reported to be mildly diuretic and to have a soothing and antiinflammatory effect on the urinary tract, as well as acting as a mild sedative. Oil made from the flowers is used soothing earache, and can be applied externally for eczema and other types of inflammatory skin conditions. These species are commonly used to treat hemorrhoids, rheumatic pain, superfacial fungal infections, wounds and diarrhoea. They are traditionally consumed as a tea to relieve abdominal pains (Tuzlaci & Erol, 1999; Sezik et al., 2001; Turker & Camper, 2002). In addition to the above mentioned common uses for the Verbascum family without referring a particular species, Verbascum lasianthum Boiss. ex Bentham flowers are reported to be used against hemorrhoids in southwest Anatolia (Tuzlaci & Erol, 1999). To evaluate the scientific basis for this practice, in vivo anti-inflammatory and antinociceptive activities were investigated. A methanolic extract of the flowers was shown to possess significant inhibitory activity in the carrageenan-induced hind paw oedema model and in p-benzoquinoneinduced writhings in mice. Through bioassay-guided fractionation and isolation procedures eight compounds, 6-0-(4"'-0-trans-p-coumaroyl}-a- Lrhamnopyranosylaucubin (1), 6-0-(4'" -O-trans-p- methoxycinnamoyl}-a-Lrhamnopyranosylaucubin (2), sinuatol (3), aucubin (4), geniposidic acid (5), catalpol (6), ajugol (7) and ilwensisaponin A (8) were isolated and their structures were elucidated by spectral techniques (Fig 12). An iridoid glucoside, aucubin (4) and a triterpenoid saponin, ilwensisaponin A (8) were found to possess significant antinociceptive and anti-inflammatory activities, per os without inducing any apparent acute toxicity or gastric damage (Kupeli et al., 2007a). Aucubin, one ofthe major iridoid glycosides of the flowers of Verbascum lasianthum, was previously shown to inhibit the TNF-a and IL-6 production in RBL-2H3 mast cells. Aucubin induced an enlarged IkE pool through inhibition ofIkE inhibited translocation ofTNFKBIRel A to nucleus, and therefore inhibited expression of inflammatory cytokines TNF -a and IL-6 (Jeong et al., 2002). Moreover, Recio et al. (1994) reported that aucubin was effective against carrageenan-induced edema. In the present study, these results supported the previous findings that aucubin is a specific inhibitor of carrageenan induced hind paw edema, which might explain its beneficial effect in the treatment of inflammatory diseases. On the other hand, aucubin derivative and catalpol have no effect against
100
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
carrageenan-induced edema, while ajugol was found to possess a weak activity in similar experimental protocols. A double bond between C-7 and C-8 is one of the most positive characters for activity, and its oxidation to an epoxy derivative leads to a remarkable decrease in anti-inflammatory activity, as occurs in catalpol (6). If an ester linking appears by acylation with an aromatic acid derivative at the sugar moiety, anti-inflammatory activity is augmented, whereas if acylation occurs directly in the monoterpenoid skeleton, this activity diminishes (Recio et al., 1994). However, in contrast with previous studies, our results showed that aucubin derivative (1) had no activity. Introduction of a hydroxyl function at C-8, as occurs in ajugol (7), decreases topical activity. The absence or presence of the extra-annual carboxyl or carboxymethyl group at C-4 is of no relevance, as is apparent result when the activity of aucubin is compared with those of geniposidic acid (5), but the conversion of a -COOH compound to its -COOMe derivative notably increases the topical activity (Recio et al., 1994). The potency ofilwensisaponin A was similar to that of indomethacin in acute anti-inflammatory experimental models and consistent, i.e. the effect was not reduced with time. It has recently been reported that other closely related saponins, verbascosaponin A and verbascosaponin, suppressed the edematous response 1 h after carrageenan injection, but this effect diminished slightly in due time (Giner et al., 2000). The mechanism of action, however, has not yet been established. It may be speculated that, the glucose molecule in the sugar residue connected to the C-3 position of the aglycone is crucial for its acute anti-inflammatory effect (Hostettmann & Marston, 1995; Gepdiremen et al., 2005). Outcome of the study, in connection with the role of aucubin and ilwensisaponin A as the active principles of V. lasianthum, it seems that they could be synergistic with other active substances in its fractions and extracts (Kupeli et al., 2007a). R1
OH
H OH
Rl 6-0-(4'" -O-trans-p-coumaroyl)H o:-L-rhamnopyranosylaucubin (2) 6-0-(4'" -O-trans-p- methoxy -cinnamoy1)- H 0:- L-rhamnopyranosylaucubin
(1)
(3) Sinuatol (4) Aucubin (5) Geniposidic acid
R2 (4'" -O-trans-p-coumaroyl)-o:-Lrhamnopyranose (4'" -O-trans-p-methoxycinnamoyl)-o:-Lrhamnopyranose 0:- L-rhamnopyranose
H H H COOH H
Fig 12. Contd.
101
Effects of Medicinal Plants in Turkey Fig 12. Contd.
~
"~~~O" H~OH
OH
(6) Catalpol H06
~ 8
H
CH3
~ 1
0
~~OH H~OH
OH
(7) Ajugol
-~q
~q
YRO~O~
Me-r--o--1I'~ RO~ RO OR
OR I"
R
OR
0
~ RO
23
I"" OR
(8) Ilwensisaponin A Fig 12. Structures of the compounds (1-8)
In a different study, saponin glycosides called ilwensisaponin A and C and iridoid glycosides known as ajugol and picroside IV (Fig 13) were isolated from the Verbascum pterocalycinum var. mutense Hub.-Mor. methanolic extract. A dose-related anti-inflammatory and antinociceptive response were obtained in the study at doses of 100 and 200 mg/kg. The results of the evaluation of the anti-inflammatory activities induced by carrageenan and PGE 1 showed that this species possesses active constituents that could diminish cyclooxygenase activitiy. No effects were seen in the 12-0tetradecanoyl-13-acetate (TPA)-induced ear edema model. Ilwensisaponin A and C could explain in part the anti-inflammatory and analgesic activities of this species. Although antinociceptive and anti-inflammatory activities of ajugol and picroside IV were not significant in statistical analysis, ilwensisaponin A and C showed notable activity without inducing any apparent acute toxicity as well as gastric damage at 200 mg/kg dose (Kiipeli Akkol et al., 2007). In a reference survey, some triterpenes have been reported
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
102
to have anti-inflammatory, cytotoxic, antitumor activities and to be chemopreventive. Papillomas in mouse skin were initiated with 390 nmol of7,12-dimethylbenz(a)anthracene and 1 week later, were promoted 2 times a week with 4.1 nmol of 12-0-tetradecanoylphorbol-13-acetate (TPA). Five saponin-related compounds used as potential anti-inflammatory agents from Verbascum songaricum were applied in the same position with 82 nmol. Saponin-related compounds effectively inhibited tumor formation in the sensitive mouse stock even when these compounds were given 1 h prior to TPA treatment. These results suggested that there was a general correlation between the anti-inflammatory and antitumor-promoting activities of saponins (Tokuda et al., 1991). Songarosaponins and their acylated derivatives, which had been previously isolated from Verbascum songaricum, have also been tested for anti-inflammatory activity using the croton oil ear model. Acylated derivatives showed excellent anti-inflammatory activity as compared to that of saponins (Anam, 2001). Giner et al. (2000) also demonstrated that verbascosaponin A and verbascosaponin, isolated from Scrophularia auriculata, significantly inhibited the mouse paw edema induced by carrageenan and ear edema induced by single and multiple doses ofTPA (Giner et al., 2000).
OH
O~O~O
H~ml~O HO
OH
~OH I"
HO H
OH
0
0
1m• OH
Ilwensisaponin C HO
7
o HO--'::::
O~O~O HO~OH OH
OH
o
Picroside IV Fig 13. Chemical structures ofisolated compounds
Effects of Medicinal Plants in Turkey
103
Several Veronica species are reported to possess application in traditional medicines worldwide for the treatment of a wide range of disorders; as expectorant in respiratory diseases or as antiscorbutic, as diuretics and for wound healing (Baytop, 1999; Harput et al., 2002). In Chinese traditional medicine, V. anagallis-aquatica L. is used for the treatment of influenza, hemoptysis, laryngopharyngitis and hernia (Su et al., 1999). During our field expeditions on Turkish folk medicine, we have recorded that aerial parts ofV. anagallis-aquatica is boiled in milk to obtain poultice and then is applied on abdomen for abdominal pain or warm aqueous extract of leaves without removing the boiled herbs is used as a bath remedy to alleviate rheumatic pain in northwest Anatolia (Fujita et al., 1995). Previously, in vitro anti-inflammatory activity of five Veronica species (V. cymbalaria, V. hederifolia, V. pectinata var. glandulosa, V. persica and V. polita) were investigated through evaluation of the inhibitory effects on nitric oxide (NO) production in lipopolysaccharide-stimulated mouse peritoneal macrophages as well as cytotoxic activity against KB epidermoid carcinoma and B16 melanoma (Harput et al., 2002). MeOH extracts offive plant materials were found to possess inhibitory effects on NO synthesis in varying degrees. MeOH extracts were further partitioned between water and chloroform, and aqueous fractions showed the activity without inducing any cytotoxicity, while chloroform fractions were cytotoxic dose-dependently. Moreover, water fractions were found to possess remarkable effect on DPPH (2,2-diphenyl-1-picryl-hydrazyl), suggesting that the inhibitory effect on NO production might be due to their radical scavenging activity. In a study performed by our group, methanolic extract of the Veronica anagallisaquatica L. plant was shown to possess significant inhibitory activity against carrageenan-induced hind paw edema model and ofp-benzoquinone-induced writhings in mice. Through bioassay-guided fractionation and isolation procedures eight compounds, aquaticoside A (1), aquaticoside B (2), aquaticoside C (3), veronicoside (4), catalposide (5), verproside (6), verminoside (7) and martynoside (8) were isolated and their structures were elucidated by spectral techniques (Fig 14). Catapol derivative iridoid glucosides, catalposide (5) and verproside (6) were found to possess potent antinociceptive and anti-inflammatory activities at 500 mg/kg dose, per os without inducing any apparent acute toxicity as well as gastric damage (Kiipeli et al., 2005). Previously, anti-inflammatory effect of several iridoid derivatives has been reported by several other authors. Recio et al. (1994) studied the anti-inflammatory activity oftwelve iridoid glycosides against two inflammation models in mice, carrageenan-induced paw edema and TPA-induced mouse ear edema. Loganic acid and two catalpol derivatives, aucubin and 6' -vanilloylcatalpol, were found to possess potent inhibitory activity on the former, while catalpol derivatives, aucubin, verbenalin and loganin, were found remarkably active on the latter model. According to
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
104
their experimental results, systemic anti-inflammatory activity ofiridoids was modest, while a higher efficacy could be observed through topic application as evidenced by TPA-edema model. They have further suggested a structure-activity relationship for topic anti-inflammatory activity of iridoids. Accordingly, OH-substitution as C 5 , unsaturation as C 7-C S' methyl substitution of carbonyl Cll' and integrity of the cyclopentane ring were essential for higher activity. Some of the results suggested in the study of Recio et al. (1994) were later supported by other authors as well; i.e. picroside II, a catalpol derivative, was found active only topically and exerted a moderate topic anti-inflammatory activity in mouse ear-swelling assay (Jia et al., 1999). This conclusion also supports the folkloric application of the plant in Turkey, where people applied boiled herbs ofV. anagallis-aquatica on their extremities, while taking bath in warm aqueous extract to alleviate rheumatic pain (Fujita et al., 1995). Furthermore, the conclusion suggested by Recio et al. (1994) reporting that a higher efficacy could be observed through topic application of iridoid glucosides than oral administration provides a further support to folkloric utilization in Turkey,
HO
o 1
10
0
I'
6'
3"
O-~o-R
~~H
-5"
OH R
H
(1) Aquaticoside A
OH
(2) Aquaticoside B
11
COOH
HO
o
0
3"
II~ -+-O~O-C~ °HO~OH -5" 1,
OH (3) Aquaticoside C
105
Effects of Medicinal Plants in Turkey Fig 14. Contd.
o I
I'
O~O~CH20H
HO~OH OH
R2
RI H H
H
(4) Veronicoside (5) Catalposide (6) Verproside
OH OH
OH
o II
7"
HO~" I" C-O
I
HO
'<:::::,
~4 5 ~3
'<:::::,8"
0
.#
5"
.
HOH2 /
9
8
10
0
OI~I' 0 CH20H OH
HO
OH
(7) Verminoside
~ HO
1'"
13'
~a'
~OH 4' 0
o 0
o
;:;H C--J-O--J I "
OO~ 3
HO
13
0H l'O~~ 3 a
3'
OH
~ 5
I
O~
OH (8) Martynoside
Fig 14. Chemical structures of isolated compounds 1-8
TAXACEAE
There are eight Taxus species and two hybrids in the world (Van Rozendall et al., 1999) and T. baccata L. (European yew) is the single representative in Turkey (Davis & Cullen, 1965). A large number of taxoids possessing different skeleton systems, as well as lignans, flavonoids, steroids and sugar derivatives were isolated from various Taxus species (Baloglu & Kingston, 1999; Parmar et al., 1999). During the course of studies on the bioactive
106
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
components, the chloroform soluble portion of ethanolic extract of the heartwood ofT. baccata afforded four taxoids; taxusin, baccatin VI, baccatin III and 1b-hydroxybaccatin I, along with five lignans; lariciresinol, taxiresinol (3'-demethyllariciresinoD, 3'-demethylisolariciresinol-9'hydroxyisopropylether, isolariciresinol and 3-demethyl isolariciresinol (Erdemoglu, 1999). Lignans are known to possess various biological activities, including antibacterial, antifungal, antiviral, antioxidant, anticancer and anti-inflammatory effects (Cho et al., 2001a). As related with the abovepresented data, a study was designed to investigate in vivo anti-inflammatory and antinociceptive activity of lignans and taxoids isolated from the chloroform-soluble portion of ethanolic extract of the heartwood of T. baccata. All the isolated lignan derivatives and taxoid compounds by Erdemoglu et al. (1999) were shown to possess significant antinociceptive activity against p-benzoquinone induced abdominal contractions, while only lignan derivatives significantly inhibited carrageenan-induced hind paw edema in mice (Kiipeli et al., 2004). These results were in accordance with the previous studies. Two of the studied lignans; lariciresinol and isolariciresinol, were reported to possess potent in vitro inhibitory effect on TNF- production, a pro-inflammatory cytokine (Cho et al., 2001b); and thus lignan derivatives in the present study might show their anti-inflammatory effects via the same mode. On the contrary, the weak anti-inflammatory effect was observed for the taxoids studied in the present study, which might be ascribed to their TNF-inducing effect when stimulated with Salmonella LPS, as reported by Kirikae et al. (2000).
THYMELAEACEAE In Traditional Chinese Medicine, several Daphne species have been frequently added in formulations prescribed against inflammatory disorders; i.e. D. genkwa to dispel edema (Zheng et al., 2006), D. acutiloba for wounds and bruises (Taniguchi et al., 1998), D. tangutica for treatment of rheumatoid arthritis (Chen et al., 2004), seeds and barks, rarely leaves, of D. mezerium in chronic rheumatism, skin diseases, gout and inflammations in the lymph tissue (Chen et al., 2004). A piece of root ofthe latter species was also recommended to stop pain in toothache by chewing (Grieve & Leyel, 1967; Schildknecht et al., 1970). Aerial parts and roots of Daphne oleo ides ssp. oleoides are used for the treatment of rheumatic pain, lumbago, and to reduce fever in Turkish folk medicine. In Inner Anatolia, the fresh stem of the plant was employed to reduce swellings in the legs of cripple animals (Baytop, 1999; Yesilada et al., 2001). In an activity screening study on a number of plant extracts used for treatment of various inflammatory conditions, D. oleoides ssp. oleoides of Turkish origin was found to be the most effective against interleukin-1ex and TNF-ex (Yesilada et al., 1997). Then, through in vitro activity-guided fractionation procedures assessing the inhibitory effects on inflammatory cytokines two diterpenes and one
Effects of Medicinal Plants in Turkey
107
coumarin derivative were isolated and chemically defined as the main principles that supported its the folkloric usage (Yesilada et al., 2001). In another study the in vivo anti-inflammatory and antinociceptive activities of D. pontica were studied. For this purpose, effects of extracts and fractions obtained through successive solvent extractions with n-hexane, diethyl ether, ethyl acetate and methanol from either the roots, leaves, stems or flowers with young leaves were investigated against carrageenan-induced hind paw edema, PGE 2-induced hind paw edema and 12-0-tetradecanoyl-13-acetate (TPA)-induced mouse ear edema models for the anti-inflammatory activity, and p-benzoquinone-induced abdominal constriction test for the antinociceptive activity assessment. Only ethyl acetate extracts ofthe roots showed significant anti-inflammatory activity on carrageenan-induced (22.732.0% inhibition) and PGE 2-induced hind paw edema (3.2-27.3% inhibition) as well as 12-0-tetradecanoyl-13-acetate (TPA)-induced mouse ear edema (47.8-43.3% inhibition) models at 50 mg/kg dose without inducing any apparent gastric lesion or acute toxicity, whereas the other extracts were shown to be ineffective. In addition to roots, ethyl acetate extracts of the stems also exhibited 19.5-29.9%; 5.3-23.9%; 36.6-28.1% inhibition on carrageenan-induced and PGE 2-induced hind paw edema as well as 12-0tetradecanoyl-13-acetate (TPA)-induced mouse ear edema models, respectively. On the other hand, none ofthe extracts showed antinociceptive activity (Kupeli et al., 2007b). TILIACEAE Silver linden, Tilia argentea Desf. ex DC. (Tiliaceae) [synonym: T. tomentosa Moench] inflorescence is a popular remedy worldwide against the symptoms of common cold, bronchitis and cough (Gruenwald et al., 2000b). Moreover, it is also claimed to be effective as a diaphoretic for feverish colds and infectious diseases where a sweating cure is needed as well as occasionally used as a stomachic, antispasmodic, diuretic and sedative, although these effects are notified to need experimental proof (Gruenwald et al., 2000). The use ofleaves as medicine is not common as that offlowers, but suggested to be employed as a diaphoretic, however the effect has not been evaluated scientifically so far. It is also reported that the Tilia flavonoids to possess some anti-edema effect, however the composition was not described (Gruenwald et al., 2000). In order to assess this information, antinociceptive and anti-inflammatory activity of the main two flavonoid glycosides; kaempferol-3, 7 -O-a-dirhamnoside and quercetin-3, 7 -O-a-dirhamnoside isolated from the leaves were investigated. Both compounds were shown to possess potent antinociceptive and anti-inflammatory activity in 50 mg! kg dose, per os, without inducing any apparent acute toxicity as well as gastric damage (Toker et al., 2004). The potent anti-inflammatory activity of different types of flavonoids was reported previously. The anti-
108
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
inflammatory activity of quercetin was reported by Simoes et al. (1988) against carrageenan-induced paw edema model at a dose of20 mg/kg i.p. and was noted a more pronounced protective effect on the earlier stages of the oedematogenic response. It is suggested that some flavonoids including quercetin blocked both the cyclooxygenase and lipoxygenase pathways of the arachidonate cascade at relatively high concentrations, while at lower concentrations only the lipoxygenase pathway (Di Carlo et al., 1999). Actually these data were supported by the conclusion that quercetin is an effective inhibitor of phospholipase A2 which catalyses the hydrolysis of phospholipids to release arachidonic acid as the precursor of the inflammatory response (Harborne, 1994). Guardia et al. (2001) studied the anti-inflammatory activity of three flavonoids; quercetin (flavonol), hesperidin (flavanone glycoside) and rutin (quercetin rutinoside) in 80 mg! kg doses using an acute/subacute inflammation models. Accordingly, rutin was found very active in suppressing the edema, nodules and ankylosis as evidenced from arthrogram scores in subacute phase, while quercetin and hesperidin were shown to possess low activity. Rotelli et al. (2003) conducted an in vivo comparative study on the anti-inflammatory activity of various flavonoids (quercetin, rutin, morin, hesperetin and hesperidin) and found that quercetin (in 75 mg/kg dose) showed a remarkable inhibitory activity on carrageeenan-induced paw edema model on acute administration (between 55 to 66% inhibition) as well as on the chronic model against adjuvant-induced arthritis model, while rutin (in 150 mg/ kg dose) was only effective against the later. Sanchez de Medina et al. (2002) pointed out that quercitrin exerts anti-inflammatory activity in trinitrobenzene sulfonic acid-induced colitis with a narrow dose-response. As described above, quercitrin is also one of the flavonoid components of silver linden leaves. Matsuda et al. (2002) reported that the kaempferol glycosides (tiliroside and astragalin) to possess potent inhibitory effect on TNF-production. However, there is no experimental study on the antiinflammatory activity of kaempferol glycosides. Detailed information on the absorption features offlavonoids have not yet to be available. Although it has been claimed that only the flavonoid aglycones be able to pass through the gut wall, this might be largely depends on the chemical structure of flavonoid (Di Carlo et al., 1999). However, once absorbed, flavonoids influence many biological functions including protein synthesis, cell proliferation differentiation and angiogenesis for the benefit of mankind (Di Carlo et al., 1999). If this hypothesis is correct, a-glycosidic bonds of the flavonoid glycosides 1 and 2 isolated by Toker et al. (2004) might be subjected to hydrolysis in the gut through a-glycosidase type enzymes to give aglycone then pass the gut wall to exert its anti-inflammatory and antinociceptive activity.
Effects of Medicinal Plants in Turkey
109
VITACEAE The leaves of Vitis vinifera (vine leaves) have been traditionally used as a food in both fresh and brined forms in Turkey. The leaves, due to astringent and haemostatic properties, are used in the treatment of diarrhoea, haemorrhage and varicose veins, and the juice of leaves has been used as an eye bath (Felicio et al., 2001). In order to evalute the biological effects of vine leaves, extracts were submitted to bioassay models such as antioxidant activity [free radical scavenging activity (1,1-diphenyl-2-picrylhydrazyl, DPPH% assay), iron (III) reductive activity (reducing power activity assay), capacity of inhibition of linoleic acid peroxidation (ferric thiocyanate and thiobarbituric acid method)], antinociceptive activity (p-benzoquinoneinduced abdominal constriction test) and anti-inflammatory activity (carrageenan-induced hind paw oedema model). All the vine leaf extracts were able to scavenge DPPH% radicals at physiological pH and did so in a concentration-dependent fashion as well as reduced iron (III) to iron (II). However none of the extracts was as effective as the positive controls ascorbic acid, BHT, and gallic acid. The aqueous extract prepared from fresh leaves showed moderate inhibition at 100 mg/kg dose on carrageenaninduced hind paw oedema and p-benzoquinone-induced abdominal constriction tests without inducing any gastric damage. The content of phenolic compounds in the extracts were also determined and hydroxycinnamic acids (e.g. caffeic acid), and flavonoids (e.g. quercetin) were verified by a reverse phase HPLC-PDA method in the extracts (Ko~ar et al., 2007).
CONCLUSIONS Ethnobotanical and ethnopharmacological surveys conducted by our research group have explicitly revealed that Turkish traditional medicines provide a vast source for the discovery of new leads to develop effective and safety agents for the treatment of diverse diseases. Detailed and programmed investigations on this subject along with clinical evaluation, derivatization and formulation studies may yield efficient products for the benefit of human health.
REFERENCES Adam, S.E.I., AI-Yahya, M.A. and AI-Farhan, A.H. (2001). Response of Najdi sheep to oral administration of Citrullus colocynthis, Nerium oleander leaves or their mixture. Small Ruminant Research, 40: 239-244. Akcos, Y., Ye~ilada, E. and Ezer, N. (1998). Anti-inflammatory activity of some Turkish Rubus species. Hacettepe University Journal of Faculty of Pharmacy, 18: 33-8. Ali-Shtayeh, M.S., Yaghmour, M.R., Faidi, Y.R., Salem, K. and AI-Nuri, M.A. (1998). Antimicrobial activity of 20 plants used in folkloric medicine in the Palestinian area. Journal of Ethnopharmacology, 60: 265-71. Afifi, F.U., Khalil, E. and Abdalla, S. (1999). Effect of isoorientin isolated from Arum palaestinum on uterine smooth muscle of rats and guinea pigs. Journal of Ethnopharmacology, 65: 173-177.
110
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Afifi, F.D., Shervingtion, A. and Darwish, R. (1997). Phytochemical and biological evaluation of Arum palaestinum. Part 1: Flavone C-glycosides. Acta Technologiae et Legis Medicamenti, 8(2): 105-110. Anam, E.M. (2001). Anti-inflammatory activity of compounds isolated from the aerial parts of Abrus precatorius (Fabaceae). Phytomedicine, 8: 24-27. Antonone, R., Simone, F., Morrica, P. and Ramundo, O. (1988). Traditional phytotherapy in the roccaonfina volcanic group, campania, southern Italy. Journal of Ethnopharmacology, 22: 295-306. Arimoto, T., Ichinose, T., Yashikawa, T. and Shibamoto, T. (2000). Effect of the natural antioxidant 2"-O-glycosylisovitexin on superoxide and hydroxyl radical generation. Food and Chemical Toxicology, 38: 849-852. Authore, G., Rastrelli, L., Lauro, M.R., Marzocco, S., Sorrentino, R., Sorrentino, D., Pinto, A. and Aquino, R. (2001). Inhibition of nitric oxide synthase expression by a methanolic extract of Crescentia alata and its derived flavonoids. Life Sciences, 70: 523-524. Aytac;, Z. and Aksoy, A. (2000). A new Sideritis species (Labiatae) from Turkey. Flora Mediterranea, 10: 181-184. Ba~er, KH.C., Honda, G. and Miki, W. (1986). Herb Drugs and Herbalists in Turkey. Instutute for the study of Languages and Cultures of Asia and Mrica. Tokyo, pp. 191. Baloglu, E. and Kingston, D.G.!. (1999). The Taxane diterpenoids. Journal of Natural Products, 62: 1448-1472. Bartram, T. (1995). Encyclopedia of Herbal Medicine. Grace, Dorsett, pp. 296. Bas, E., recio, M.C., Giner, R.M., manez, S., Cerda-Nicolas, M. and Rios, J.L. (2006). Antiinflammatory activity of 5-0-demethylnobilewtin, a polymethoxyflavone isolated from Sideritis tragonganum. Planta Medica, 72: 136-42. Baytop, T. (1999). Therapy with plants in Turkey (Past and Present), 2nd Edition, Nobel Medical Book House, Istanbul. Bennini, B., Chulia, A.J., Kaouadji, M. and Delage, C. (1993). Phytochemistry of the Ericaceae 4. (2R,3R)-Dihidroflavonol aglycone and glicosides from Erica cinerea. Phytochemistry, 33: 1233-1236. Blumenthal, M., Busse, W.R., Goldberg, A., Gruenwald, J., Hall, T., Klein, S., Riggins, C.W., Bussing, A. and Schweizer, K (1998). Effects ofphytopreparation from Helleborus niger on immnocompetent cells in uaro. Journal of Ethnopharmacology, 59: 139-146. Bourdy, G., de Michel, L.R.C. and Roca-Coulhard, A. (2004). Pharmacopoeia in a shamanistic society: the Izoceno-Guarani (Bolivian Chaco). Journal of Ethnopharmacology, 91: 189-208. Brandi, M.L. (1997). Natural and synthetic isoflavones in the prevention and treatment of chronic diseases. Calcifwd Tissue International, 61: S5-S8. Bremness, L. (1995). The Complete Book of Herbs. Dorling Kindersley, London, D.K British Herbal Pharmacopeia, (1987). British Herbal Medicine Association, D.K Budzianowski, J., Pakulski, G. and Robak, J. (1991). Studies on antioxidative activity of some C-glycosyl flavones. Polish Journal of Pharmacolology and Pharmacy, 43: 395401. Bueno, N.R., Castilho, R.O., da Costa, R.B., Pott, A., Pott, V.J., Scheidt, G.N. and da SilvaBatista, M. (2005). Medicinal plants used by the Kaiwo and Guarani indigenous populations in the Caarapo Reserve, Mato Grosso do SuI, Brazil. Acta Botanica Brasil, 19: 39-44. Capasso, F., Mascolo, N., Autore, G., de Simone, F. and Senatore, F. (1983). Antiinflammatory and analgesic activity in alcoholic extract of Tamus communis L. Journal of Ethnopharmacology, 8: 321-325. Carini, M., Aldini, G., Furlanetto, S., Stefani, R. and Facino, R.M. (2001). LC coupled to ion-trap MS for the rapid screening and detection of polyphenol antioxidants from Helichrysum stoechas. Journal of Pharmacy and Biomedical Analysis, 24: 517-526. Carlson, G.G. and Volney, H.J. (1940). Some notes on uses of plants by the Comanche Indians. Papers of the Michigan Academy of Scwnce, Arts and Letters, 25: 517-542. Chancellor, P.M. (1985). Illustrated Handbook of the Bach Flower Remedies, C.W. Daniel Co. Ltd., Essex, England.
Effects of Medicinal Plants in Turkey
111
Chen, J., Liu, X. and Si, Y.P. (2004). Determination of daphnetin in Daphne tangutica and its medicinal preparation by liquid chromatography. Analytica Chimica Acta, 523: 2933. Chiej, R. 1984. Encyclopaedia of MedIcinal Plants. MacDonald Press, London. Cho, J.y', Park, J., Kim, P.S., Yoo, E.S., Baik, KU. and Park, M.H. (2001a). Savinin, a lignan from Pterocarpus santalinus inhibits tumor necrosis factor-alpha production and T cell proliferation. BiologIcal and Pharmaceutical Bulletin, 24: 167-171. Cho, J.Y., Kim, A.R. and Park, M.H. (2001b). Lignans from the rhizomes ofCoptisjaponica differentially act as anti-inflammatory principles. Planta Medica, 67: 312-16. Choi, J.H, Kim, D.Y., Yoon, J.H., Youn, H.Y., Yit, J.B., Rheet, H.I., Ryut, KH., Jung, K, Hant, C.K and Chot, Y.B. (2002). Effect of SKI 306X, a new herbal agent, on proteoglycan degradation in cartilage explant culture and collagenase-induced rabbit osteoarthritis model. Osteoarthr. Cartil., 10: 471-478. Chosson, E., Chaboud, A, Chulia, AJ. and Raynoud, J. (1998). Dihydroflavonol glycosides from Rhododendron ferrugmeum. Phytochemistry, 49: 1431-1433. Chulia, A., Benini, B., Kaouadji, M., Allais, D.P. and Delage, C. (1995), Two flavonol conjugates from Erica cmerea. Journal of Natural Products, 58: 560-563. Cioffi, G., Morales-Escobar, L., Braca, A and De Tommasi, N. (2003), Antioxidant chalcone glycosides and flavanones from Maclura (Chlorophora) tinctoria. Journal of Natural Products, 66: 1061-1064. Coffey, T. (1993). The history and folklore of North American wild flowers. Houghton Mifflin Company Press, New York, USA. Corra, R., Pereira, M.A., Buffon, D., Dos Santos, L., Cechinel, Filho V., Santos, A.R. and Nunes, R.J. (2001). Antinociceptive properties of chalcones. Structure-activity relationships. Archive der Pharmazie, 334: 332-334. Crowden, R.K, Harborne, J.B. and Heywood, V.H. (1969). Chemosystematics of the Umbelliferae-a general survey. Phytochemistry, 8: 1963-1984. Davis, P.H. (1982). Flora of Turkey and the Esat Aegean Islands, Vol. V. University Press Edinburgh. Davis, P.H. and Cullen, J. (1965) Taxus L., In: "Flora of Turkey and the East Aegean Islands" (Davis, P.H., Ed.), vol. 1, University Press, Edinburgh, pp. 565. Davis, P.H. and Cullen, J. (1984). Lycopodium L. In: Flora of Turkey and the east Aegean islands, Ed. Davis PH, volume 1, pp. 88-90, Edinburg University Press, Edinburg, UK Deliorman-Orhan, D., AsIan, M., Aktay, G., Ergun, E., Yesilada, E. and Ergun, F. (2003). Evaluation ofhepatoprotective effect of Gentiana olivieri herb on subacute administration and isolation of active principle. Life Scences, 72: 2273-2283. Deliorman Orhan, D., Hartevioglu, A, Kupeli, E. and Yesilada, E. (2007). In vivo antiinflammatory and antinociceptive activity of the crude extract and fractions from Rosa can ina L. Fruits. Journal of Ethnopharmacology, 112: 394-400. Deliorman Orhan, D., Kupeli, E., Yesilada, E. and Ergun, F. (2006). Anti-inflammatory and antinociceptive activity of flavonoids isolated from Viscum album ssp. album. Zeitschrift fur Naturforschung, 61c: 26-30. Di Carlo, G., Mascolo, N., Izzo, A.A and Capasso, F. (1999). Flavonoids, old and new aspects of a class of natural therapeutic drugs. Life Sciences, 65: 337-353. Didry, N., Dubreuil, L. and Pinkas, M. (1993). Microbiological properties of protoanemonin isolated from Ranunculus bulbosus. Phytotherapy Research, 7: 21-24. Diena, M., Rosa, A, Casu, V., Cottiglia, L., Bonsignore, L. and Dessi, M.A (2003). Chemical composition and antioxidant activity of extracts from Daphne gnidium L. Journal of the American Oil Chemists Society, 80: 65-70. Dimas, K, Demetzos, C., Mitaku, S., Marselos, M., Tzavaras, T. and Kokkinopoulos, D. (2000). Cytotoxic activity of kaempferol glycosides against human leukemic cell lines in vitro. Pharmacological Research, 41: 85-88. Dufour, D., Pichette, A, Mshvildadze, V., Bradette-Hebert, M.E., Lavoie, S., Longtin, A, Laprise, C. and Legault, J. (2007). Antioxidant, anti-inflammatory and anticancer activities of methanolic extracts from Ledum groenlandicum Retzius. Journal of Ethnopharmacology, 111: 22-28.
112
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Duman, H. (2000a). Lamium L. In: Gtiner, A., Ozhatay, N., Ekim, T., Ba~er, KH.C. (Eds), Flora of Turkey and the East Aegean Islands, vol. 11, (Suppl. 2). University Press, Edinburgh, UK, pp. 199-200. Duman, H. (2000b). Sideritis L., In: "Flora of Turkey and The East Eagean Islands" (supplement II) (Eds. A. Gimer, N. Ozhatay, T. Ekim ve KH.C. Ba~er) 11: 201-204, University Press, Edinburgh. Elvin-Lewis, M., Hall, J.B., Adut-Tutu, M., Afful, Y., Asante-Appiah, K. and Lieberman, D. (1980). The dental health of chewing stick users in Southern Ghana: preliminary findings. Journal of Preventive Dentistry, 6: 151-159. Elvin-Lewis, M. and Lewis, W.H. (1983). The dental use of plants in Amazonia. Odontostomalogique Tropicale, 6: 178-187. Erdemoglu, N. (1999). "Researches on Taxane-type compounds of Taxus baccata L. growing in Turkey", Ph.D. Thesis, Gazi University Institute of Health Sciences, Ankara. Erdemoglu, N., Kupeli, E. and Yesilada, E. (2003). Anti-inflammatory and Antinociceptive Activity Assessment of Plants Used as Remedy in Turkish folk Medicine. Journal of Ethnopharmacology, 89: 123-29. Felicio, J.D., Santos, R.S. and Gonzalez, E. (2001). Chemical constituents from Vitis vinifera (Vitaceae). Arg. Inst. Biol., 68: 47-50. Ford-Hutchinson, A.W., Bray, M.A, Cunningham, F.M., Davidson, E.M. and Smith, M.J.H. (1981). Isomers of leukotriene B possess different biological potencies. Prostaglandins, 21: 143-151. Fujiki, H., Suganuma, M., Kurusu, M., Okabe, S. Imayoshi, Y., Taniguchi, S. and Yoshida, T. (2003). New TNF-a releasing inhibitors as cancer preventive agents from traditional herbal medicine and combination cancer prevention study with EGCG and sulindac or tamoxifen. Mutation Research, 523-524: 119-125. Fujita, T., Sezik, E., Ye~ilada, E., Tabata, M., Ye~ilada, E., Honda, G., Takeda, Y., Tanaka, T. and Takaishi, Y. (1995). Traditional Medicine in Turkey VII. Folk medicine in Middle and West Black Sea regions. Economic Botany, 49: 406-422. Fukuda, K, Hibiya, Y., Mutoh, M., Koshiji, M., Akao, S. and Fujiwara, H. (1999). Inhibition by berberine of cyclooxygenase-2 transcriptional activity in human colon cancer cells. Journal of Ethnopharmacology, 66: 227-233. Gebrie, E., Makonnen, E., Debella, A. and Zerihun, L. (2005). Phytochemical screening and pharmacological evaluations for the antifertility effect of the methanol root extract of Rumex steudelii. Journal of Ethnopharmacology, 96: 139-143. Gepdiremen, A., Mshvildadze, V., Suleyman, H. and Elias, R. (2005). Acute antiinflammatory activity of four saponins isolated from ivy; Alpha-hederin, hederasaponinC, hederacolchiside-E and hederacolchiside-F in carrageenan-induced rat paw edema. Phytomedicine, 12: 440-444. Getie, M., Gebre-Mariam, T., Rietz, R, Hahne, C, Hushcka, C., Schmidtke, Abate, A. and Neubert, RH.H. (2003). Evaluation of anti-microbial and anti-inflammatory activities of the medicinal plants Dodonaea viscosa, Rumex nervosus and Rumex abyssinicus. Fitoterapia, 74: 139-143. Ghosh, L., Gayen, J.R, Murugesan, T., Sinha, S., Pal, M. and Saha, B.P. (2003). Evaluation of purgative activity of roots of Rumex nepalensis. Fitoterapia, 74: 372-374. Gil, B., Sanz, M.J., Ferrandiz, M.C., Bustos, M.P., Gunasegaran, R and Alcaraz, M.J. (1994). Effect of flavonoids on Naja naja human recombinant synovial phospholipases ~ and inflammatory responses in mice. Ltfe Sciences, 54: 333-338. Giner, R-M., Villalba, M.L., Recio, M.C., Manez, S., Cerda-Nicolas, M. and Rios, J.L. (2000). Anti-inflammatory glycoterpenoids from Scrophulana auriculata. European Journal of Pharmacology, 389: 243-252. Girzu, M., Carnat, A., Privat, A.M., Fialip, J., Carnat, A.P. and Lamaison, J.L. (1998). Sedative effect of walnut leaf extract and juglone, an isolated constituents. Pharmaceutical Biology, 36: 280-286. Grieve, M. (1994). A Modern Herbal (Leyel, C.F., Ed.), Tiger Books International, London. Grieve, M. and Leyel, C.F. (1967). A modern herbal, the medicinal, culinary, cosmetic and economic properties, cultivation and folklore of herbs, grasses, fungi; shrubs and trees. Vol. II, Herner Publishing Co., New York.
Effects of Medicinal Plants in Turkey
113
Grieve, M. (1994). A Modern Herbal. Tiger Books International, London pp. 207-208. Godoy, A., de Heras, B., Vivas, J.M. and Villar, A. (2000). Anti-inflammatory properties of a lipid fraction obtained from Sideritis javalambrensis. Biological and Pharmaceutical Bulletin, 23: 1193-1997. Gruenwald, J., Brendler, T. and Jaenicke, C. (2000al. PDR for Herbal Medicines. 2 nd Edition. Medical Economics Company. New Jersey, pp. 729-733. Gruenwald, J., Brendler, T. and Jaenicke, C. (2000b). PDR for Herbal Medicines. 2 nd ed., Thompson-Physicians' Desk Reference, pp. 477-8. Guardia, T., Rotelli, A.E., Juarez A.O. and Pelzer, L.E. (2001). Anti-inflammatory properties of plant flavonoids. Effects of rutin, quercetin and hesperidin on adjuvant arthritis in rat. II Farmaco, 56: 683-687. Guo, Q., Rimbach, G., Moini, H., Weber, S. and Packer, L. (2002). ESR and cell culture studies on free radical-scavenging and antioxidant activities ofisoflavonoids. Toxicology, 179: 171-180. Halliwell, B. and Chirica, S. 1993. lipid peroxidation: its mechanism, measurment, and significance. American Journal of Clinical Nutrition, 57: 715-725. Hamlyn, P. (1969). In: "The Marshall Cavendish, Encyclopedia of Gardening", vol. 19, Garrod and Lofthouse International, London, pp. 2034. Harborne, J.B. (1988). The Flavonoids: Advances in Research Since (1980) Chapman & Hall, London. Harborne, J.B. (1994). The Flavonoids: Advances in Research since 1986. Chapman & Hall, London. Harput, U.S., Saracoglu, I., Inoue, M. and Ogihara, Y. (2002). Anti-inflammatory and cytotoxic activities of five Veronica species. Biological and Pharmaceutical Bulletin, 25: 483-486. Herencia, F., Ferrandiz, L.M., Ubeda, A., Guillen, I., Dominguez, J.N., Charris, J.E., Lobo, G.M. and Alcaraz, M.J. (1999). Novel anti-inflammatory chalcone derivatives inhibit the induction of nitric oxide synthase and cyclooxygenase-2 in mouse peritoneal macrophages. FEBS Letters, 453: 129-134. Holdsworth, D.K and Giheno, J. (1975). A preliminary survey of highland medicinal plants. Science in New Guinea, 3: 191-198. Hom-Ross, P.L., John, E.M., Canchola, A.J., Stewart, S.L. and Lee, M.M. (2003). Phytoestrogen intake and endometrial cancer risk. Journal of National Cancer Institute, 95: 1158-1164. Honda, G., Yesilada, E., Tabata, M., Sezik, E., Fujita, T., Takeda, Y., Yoshihisa, T. and Toshihiro, T. (1996). Traditional Medicine in Turkey VI. Folk medicine in West Anatolia: AiYon, Kiitahya, Denizli, Mugla, Aydin provinces. Journal of Ethnopharmacology, 53: 75-87. Hosseinzadeh, H., Haddadkhodaparast, M.H. and Arash, A.R. (2003). Antinociceptive, antiinflammatory and acute toxicity effects of Salvia leriifolia Benth. seed extract in mice and rats. Phytotherapy Research, 17: 422-425. Hostettmann, K and Marston, A. (1995). Saponins. Cambridge University Press, Cambridge, UK Hu, C.-Q., Chang, J.-J. and Lee, K-H. (1990). Antitumor Agents, 115. Seselidiol, a new cytotoxic polyacetylene from Seseli mairei. Journal of Natural Products, 53: 932-935. Ikram, M., Ehsanul, M. and Warsi, S.A. (1966). Alkaloids of Berberis lycium. Royle-I, Pakistan Journal of Science and Industrial Research, 9: 343-6. Iscan, G., Kirimer, N., Kurkcuoglu, M., Kupeli, E., Arabaci, T. and Baser, KH.C. (2006). Biological activity and composition of the essential oils of Achillea schischkinii Sosn. and Achillea aleppica DC. subsp. aleppica. Journal of Agricultural and Food Chemistry, 54: 170-173. Jeong, H.J., Koo, H.N., Na, H.J., Kim, M.S. Hong, S.H., Eom, J.W., Kim, K-S., Shin, T.Y. and Kim, H.M. (2002). Inhibition of the TNF-a and IL-6 production by aucubin through blockade of NF-Jd3 activation in RBL-2H3 mast cells. Cytokine, 18(5): 252-259. Jouad, H., Maghrani, M. and Eddouks, M. (2002). Hypolycaemic effect of Rubus fructicosis L. and Globularia alypum L. in normal and streptozotocin-induced diabetic rats. Journal of Ethnopharmacology, 81: 351-356.
114
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
Keys, D.J. (1985). Chinese Herbs, The Botany, Chemistry and Pharmacodynamics, Third Press, Charles E. Tutle Company, Tokyo, Japan pp. 159-160. Kim, S.Y., Moon, T.C., Chang, H.W., Son, KH., Kang, S.S. and Kim, H.P. (2002). Effects of Tanshinone I isolated from Salvia miltiorrh!za Bunge on arachidonic acid metabolism and In vivo inflammatory responses. Phytotherapy Research, 16: 616-620. Kirikae, T., Ojima, I., Fuero-Oderda, C., Lin, S., Kirikae, F., Hashimoto, M. and Nakano, M. (2000). Structural significance of the acyl group at the C-10 position and the A ring of the taxane core of paclitaxel fro inducing nitric oxide and tumor necrosis factor production by murine macrophages. FEBS, 478: 221-226. Kintzios, S.E. (2000). Sage The Genus Salvia. Harwood Academic Publishers, ix-xii. Ko, F.N., Chu, C.C., Lin, C.N., Chang, C.C. and Teng, C.M. (1998). Isoorientin-6"-O-glucoside, a water-soluble antioxidant isolated from Gentwna arisanensis. Biochimica et Biophysica Acta, 1389: 81-90. Ko~ar, M. (1999). Tiirkiye'de yeti~en Berberis L. tiirlerinin alkaloitleri (Alkaloids of Berberis species growing in Turkey). Ph.D. Thesis, Anadolu University, Institute of Health Sciences, Eski~ehir (Turkey). Ko~ar, M., Kiipeli, E., Malyer, H., Uyla~er, V., Tiirkben, C. and Ba~er, K.H.C. (2007). Antioxidant, anti-inflammatory and antinociceptive activities and composition of Vitis vinifera L. (cv. Sultani Qekirdeksiz) leaves and its ethnic product. Journal of Agricultural and Food Chemistry, 55: 4596-4603. Kiilip, J. (1997). A preliminary survey of traditional medicinal plants in the west coast and interior of Sabah. Journal of Tropical Forest Science, 10: 271-274. Kumarasamy, Y., Cox, P.J., Jaspars, M., Nahar, L. and Sarker, S.D. (2002). Screening seeds of Scottish plants for antibacterial activity. Journal of Ethnopharmacology, 83: 73-77. Kiipeli, E., Tatli, I., Akdemir, Z.S. and Ye~ilada, E. (2007a). Bioassay-guided isolation of anti-inflammatory and antinociceptive glycoterpenoids from the flowers of Verbascum lasianthum Boiss. ex Bentham. Journal of Ethnopharmacology, 110: 444-450. Kiipeli, E., Tosun, A. and Ye~ilada, E. (2007b). Assesment of anti-inflammatory and antinociceptive activities of Daphne pontica L. (Thymelaeaceae). Journal of Ethnopharmacology, 113: 332-337. Kiipeli, E., Orhan, I., Toker, G. and Yesilada, E. (2006). Anti-inflammatory and antinociceptive potential of Maclura pomifera (Rafin.) Schneider fruit extracts and its major isoflavonoids, scandenone and auriculasin. Journal of Ethnopharmacology, 107: 169174. Kupeli, E., Orhan, I. and Ye~ilada, E. (2007c). Evaluation of some plants used in Turkish folk medicine for their anti-inflammatory and antinociceptive activities. Pharmaceutical Biology, 45(7): 1-9. Kiipeli Akkol, E., Gager, F., Ko~ar, M. and Ba~er, KH.C. (2008a). Anti-inflammatory and antinociceptive activities of Salvia halophila and Salvia virgata from Turkey. Food Chemistry, 108: 942-949. Kiipeli Akkol, E., Tatli, I.I. and Akdemir, Z.S. (2007). Antinociceptive and anti-inflammatory effects of saponin and iridoid glucosides from Verbascum pterocalycinum var. mutense Hub.-Mor. Zeitschrift fur Naturforschung, 62c: 813-820. Kiipeli Akkol, E., Yalcin, F.N., Kaya, D., Qal)§, I., Ersoz, T. and Ye~ilada, E. (2008b). In vivo anti-inflammatory and antinociceptive actions of some Lamium species. Journal of Ethnopharmacology, (in press). Kiipeli Akkol, E., Ye~ilada, E. and Giivenr;, A. (2008c). Valuation of anti-inflammatory and antinociceptive activities of Erica species native to Turkey. Journal of Ethnopharmacology, 116: 251-257. Kiipeli, E., Erdemoglu, N. Ye~ilada, E. and Sener, B. (2003). Anti-inflammatory and antinociceptive activity of taxoids and lignans from the Heartwood of Taxus baccata L. Journal of Ethnopharmacology, 89: 123-29. Kiipeli, E., Harput, U.S., Varel, M., Yesilada, E. and Saracoglu, I. (2005). Bioassay-guided isolation of iridoid glucosides with antinociceptive and anti-inflammatory activities from Veronica anagallis-aquat!ca L. Journal of Ethnopharmacology, 102: 170-176.
Effects of Medicinal Plants in Turkey
115
Kupeli, E., Karlal, M., AsIan, S. and Yesilada, E. (2006a). Comparative evaluation of the anti-inflammatory and antinociceptive activity of Turkish Eryngium species. Journal of Ethnopharmacology, 107: 32-37. Kupeli, E., Ko~ar, M., Yesilada, E. and Ba~er, K.H.C. (2002). A comparative study on the anti-inflammatory, analgesic and febrifuge effects of isoquinoline alkaloids from the roots of Turkish Berberis Species. Life Sciences, 72: 645-57. Kupeli, E., Orhan, I., Kusmenoglu, S. and Yesilada, E. (2007a). Evaluation of antiinflammatory and antinociceptive activity of five Anatolian Achillea species. Turkish Journal of Pharmaceutical Sciences, 4: 89-100. Kupeli, E., Sahin, F.P., Calis, 1., Yesilada, E. and Ezer, N. (2007b). Phenolic compounds of Sideritis ozturkii and their in vivo anti-inflammatory and antinociceptive activities. Journal of Ethnopharmacology, 112: 356-360 (2007). Kupeli, E., Sahin, F.P., Calis, 1., Yesilada, E. and Ezer, N. (2007c). In vivo anti-inflammatory and antinociceptive activity evaluation of phenolic compounds from Sideritis stricta. Zeitschrift fur Naturforschung, 62c: 519-525 (2007). Kupeli, E., Tatli, 1.1., Akdemir, Z.S. and Yesilada, E. (2007d). Estimation of antinociceptive and anti-inflammatory activity on Geranium pratense subsp. finitimum and its phenolic compounds. Journal of Ethnopharmacology, 114: 234-240. Ktipeli, E., Tosun, A. and Bahadir, O. (2006b). Evaluation of anti-inflammatory and antinociceptive activities of Helichrysum Gaertner species (Asteraceael. TurkIsh Journal of Pharmaceutical Sciences, 3: 141-149. Kupeli, E., Tosun, A. and Yesilada, E. (2006c). Anti-inflammatory and antinociceptive activities of Seseli L. species (Apiaceae) growing in Turkey. Journal of Ethnopharmacology, 104: 310-314. Kupeli, E. and Yesilada, E. (2007). Flavonoids with anti-inflammatory and antinociceptive activity from Cistus laurifolius leaves through bioassay-guided procedures. Journal of Ethnopharmacology, 112: 524-530. Jakovlev, V.Y. and Schlichtegroll, A. (1969). On the inflammation inhibitory effect of (-)-a-Bisabolol, an essential oil component of Chamomilla oil. ArzneimittelforschungDrug Research, 19: 615-616. Launert, E. (1981). Edible and MedIcinal Plants. Hamlyn Press, London. Lawrence, G.H.M. (1969). In: "Taxonomy of Vascular Plants", The Macmillan Company: New York, pp. 642-646. Lawrence, G.H.M. (989). Taxonomy of Vascular Plants, Mac Millan Publishing Co., pp. 337-338, New York, USA. Leporatti, M.L. and Ivancheva, S. (2003). Preliminary comparative analysis of medicinal plants used in the traditional medicine of Bulgaria and Italy. Journal of Ethnopharmacology, 87: 123-42. Lev, E. (2002). Reconstructed materia medica of the Medieval and Ottoman aI-Sham. Journal of Ethnopharmacology, 80: 167-179. Li, C.J., Wang, L.Q., Chen, S.N. and Qin, G.W. (2000). Diterpenoids from the fruits of Rhododendron mollei. Journal of Natural Products, 63: 1214-1217. Li, R.W., Lin, G.D., Myers, S.P. and Leach, D.N. (2003l. Anti-inflammatory activity of Chinese medicinal vine plants. Journal of Ethnopharmacology, 85: 61-67. Li, R.W., Myers, S.P., Leach, D.N., Lin, G.D. and Leach, G. (2002). A cross-cultural study: anti-inflammatory activity of Australian and Chinese plants. Journal of Ethnopharmacology, 85: 1-8. Liang, Y.C., Huang, Y.T., Tsai, S.H., Lin-Shiau, S.Y., Chen, C.F. and Lin, J.K. (1999). Suppression of inducible cyclooxygenase and inducible nitric oxide synthase by apigenin and related flavonoids in mouse macrophages. Carcinogenesis, 20: 1945-1952. Linke, S., Dufter, C., Kerek, F., Jung, M., Watzlik, A., Jung, T., Opelz, G. and Terness, P. (1998). Functional characterization of HP12: a novel immunosuppressant purified from Helleborus species. Transplantation Proceedings, 30: 4106-4107. Liu, F. and Ng, T.B. (20001. Antioxidative and free radical scavenging activities of selected medicinal herbs. Life Science, 66: 725-735.
116
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Lourens, A.C.U., Reddy, D., Ba~er, KH.C., Viljoen, A.M. and Van Vuuren, S.F. (2004). In vitro biological activity and essential oil composition of four indigenous South African Helichrysum species. Journal of Ethnopharmacology, 95: 253-258. Mabey, R. (1979). Plants with a Purpose. Fontana Presshouse, London. Mantle, D., Eddeb, F. and Pickering, A.T. (2000). Comparison of relative antioxidant activities of British medicinal plant species In vitro. Journal of Ethnopharmacology, 72: 47-51. Mascolo, N., Autore, G. and Capasso, F. (1987). Local anti-inflammatory activity of Tamus communis. Journal of Ethnopharmacology, 19: 81-84. Marquina, M.A., Corao, G.M., Araujo, L., Buitrago, D. and Sosa, M. (2002). Hyaluronidase inhibitory activity form the polyphenols in the fruit of blackberry (Rubus fruticosus B.), Fitoterapia 73: 727-729. Matkowsi, A. and Piotrowska, M. (2006). Antioxidant and free radical scavenging activities of some medicinal plants from Lamiaceae. Fitoterapia, 77: 346-353. Matsuda H., Ninomiya, K, Shimoda, H. and Yoshikawa, M. (2002). Hepatoprotective principles from the flowers of Tilia argentea (Linden): Structure requirement of tiliroside and mechanism of action. Bioorganic and Medicinal Chemistry, 10: 707-712. Middleton, E., Kandaswami, C. and Theoharides, T.C. (2000). The effects of plant flavonoids on mammalian cells: Implications for inflammation, heart disease and cancer. Pharmacological Reviews, 52: 673-751. Middleton, E. Jr. (1998). Flavonoids in the Living System, Plenum Press, New York. Mill, R.R. (1982). Lamwm L. In: Davis, P.H. (Ed.), Flora of Turkey and the East Aegean Islands, vol. 7. Edinburgh University Press, Edinburgh, UK, pp. 126-148. Miller, A.G. (1982). Viscum album L. In: Davis, P.H. (Ed.), Flora of Turkey and the East Aegean Islands, vol. 7. Edinburgh University Press, Edinburgh, pp. 547. Mouhajir, F., Hudson, J.B., Rejdali, M. and Towers, G.H.N. (2001). Multiple antiviral activities of endemic medicinal plants used by Berber people of Morocco. Pharmaceutical Biology, 39: 364-374. Mun'im, A., Negishi, O. and Ozawa, T. (2003). Antioxidative compounds from Crotalaria sessiliflora. Bioscience Bwtechnology and Biochemistry, 67: 410-414. Murakami, C., Ishijima, K, Hirota, M., Sakaguchi, K, Yoshida, H. and Mizushina, Y. (2002). Novel anti-inflammatory compounds from Rubus sieboldii, triterpenoids, are inhibitors of mammalian DNA polymerases. Biochimica et Bwphysica Acta, 1596: 193-200. Murray, T.M. (1995), The Healing Power of Herbs, 2nd edn. Prima, Rocklin, CA, pp. 253260. Najid, A., Simon, A., Cook, J., Chable-Rabinovitch, H., Delage, C., Chulia, A.J. and Rigaud, M. (1992). Characterization ofursolic acid as lipoxygenase and cyclooxygenase inhibitor using macrophages, platelets, and differentiated HL60 leukemic cells. FEBS, 299: 213217. Okuyama, E., Okamoto, Y., Yamazaki, M. and Satake, M. (1996). Pharmacologically active components of a Peruvian medicinal plant, huanarpo (Jatropha cilliata). Chem. Pharm. Bull., 44: 333-336. Olinescu, A., Hritescu, S., Poliopol, M. and Agache, F. (1986). Effect of Boicil® on some immunocompetent cells 1. In vitro modulation of the human peripheral blood lymphocyte function. Archieves of Roumanian Pathologw and Experimental Microbiology, 45: 33-45. Orhan, 1., Kiipeli, E., AsIan, M., Kartal, M. and Yesilada, E. (2006). Bioassay-guided evaluation of anti-inflammatory and antinociceptive activities of pistachio, Pistacia vera L. Journal of Ethnopharmacology, 105: 235-240. Orhan, 1., Kiipeli, E., Sener, B. and Yesilada, E. (2007a). Appraisal of anti-inflammatory potential of the clubmoss Lycopodium clavatum L. Journal of Ethnopharmacology, 109: 146-150. Orhan, 1., Kiipeli, E., Terzioglu, S. and Yesilada, E. (2007b). Bioassay-guided isolation of kaempferol-3-0-/3-D-galactoside with anti-inflammatory and antinociceptive activity from the aerial part of Calluna vulgaris L. Journal of Ethnopharmacology, 114: 32-37. Ozaydin, S., Dirmenci, T., Tiimen, G. and Ba~er, KH.C. (2006). Plants used as analgesic in the folk medicine Turkey. In: Proceedings of the 4th International Congress of Ethnobotany (ICEB 2005), Ertug, F. (ed), Ege University Publications, pp. 167-171.
Effects of Medicinal Plants in Turkey
117
Paduch, R., W6jciak-Kosior, M. and Matysik, G. (2007). Investigation of biological activity of Lamil albi flos extracts. Journal of Ethnopharmacology, 110: 69-75. Parmar, V.S., Jha, A, Bisht, KS., Taneja, P., Singh, S.K, Kumar, A, Raijni Jain, P. and Olsen, C.E. (1999). Constituents of Yew trees. Phytochemistry, 50: 1267-1304. PDR for Herbal Medicines, 2000. Second Edition, Montvale, New Jersey, pp. 769. Panizzi, L., Caponi, C., Catalano, S., Cioni, P.L. and Morelli, I. (2002). In vltra antimicrobial activity of extracts and isolated constituents of Rubus ulmifolius. Journal of Ethnopharmacology, 79: 165-168. Pelzer, L.E., Guardia T., Juarez A.O. and Guerreiro, E. (1998). Acute and chronic antiinflammatory effects of plant flavonoids. Il Farmaco, 53: 421. Pieroni, A, Nebel, S., Quave, C., Munz, H. and Heinrich, M. (2002). Ethnopharmacology of Liakra: Traditional weedy vegetables of the Arbereshe of the Vulture Area in Southern Italy. Journal of Ethnopharmacology, 81: 165-185. Pimenov, M.G. and Leonov, M.V. (1993). In: "The Genera of the Umbelliferae", Whitstable Litho: Whitstable, Kent. Rauha, J.P. (2001). The research for biological activity in Finnish plant extracts containing phenolic compounds. In: PhD Thesis at the Pharmacognosy Department of Pharmacy Faculty of Science University of Helsinki, pp. 31-45. Rauha, J.P., Remes, S., Heinonen, M., Hopia, A.I., Kahkonen, M., Kujala, T.S., Pihlaja, K, Vuorela, H.J. and Vuorela, P. (2000). Antimicrobial effects of Finnish plant extracts containing flavonoids and other phenolic compounds. International Journal of Food Microbiology, 56: 3-12. Recio, M.C., Giner, R.M., Manez, S. and Rios, J.L. (1994). Structural considerations on the iridoids as anti-inflammatory agents. Planta Medica, 60: 232-4. Reyes Ruiz, M., Martin-Cordero, C., Ayuso Gonzalez, M.J., Toro Sainz, M.V. and Alarcon de la Lastra, C. (1996). Antiulcer activity in rats by flavonoids of Erica andevalensis Cabezudo-Rivera. Phytotherapy Research, 10: 300-303. Rezaeipoor, R, Sanei, R.Z. and Kamalinejad, N., (2000). Effects of Istatis cappadoclca on humoral immune responses. Fitoterapia, 71: 14-18. Rouf, AS.S, Islam, M.S. and Rahman, M.T. (2003). Evaluation of antidiarrhoeal activity Rumex maritimus root. Journal of Ethnopharmacology, 84: 307-310. Rotelli, AE., Guardia, T., Juarez, AO., Rocha, N.E. and Pelzer, L.E. (2003). Comparative study of flavonoids in experimental models of inflammation. Pharmacological Research, 48: 601-606. Samuelsson, B., Goldyne, M., Granstrom, E., Hamberg, M., Hammarstrom, S. and Malmsten, C. (19781. Prostaglandins and tromboxanes. Annual Reviews of Biochemistry, 47: 997-1029. Sanches de Medina, F., Vera, B., Galvez, J. and Zarzuelo, A (2002). Effect of quercetin on the eraly stages of hapten induced colonic inflammation in the rat. Life Sciences, 70: 3097-3108. Southwell, LA. and Tucker, D.J. (1993). Protoanemonin in Australian Clematis. Phytochemistry, 33: 1099-1102. Schildknecht, V.H., Edelmann, G. and Maurer, R. (1970). Zur chemie des mezereins, des entziindlichen und cocarcinogenen giftes aus dem seidelbast Daphne mezereum. Chemiker-Zeitung, 94: 347-355. Sezik, E., AsIan M., Yesilada, E. and Ito, S. (2005). Hypoglycaemic activity of Gentiana olivieri and isolation of the active constituent through Bioassay-guided fractionation techniques. Llfe Sciences, 76: 1223-38. Sezik, E., Tabata, M., Yesilada, E., Honda, G., Goto, K and Ikeshiro, Y. (1991). Traditional medicine in Turkey 1. Folk medicine in Northeast Anatolia. Journal of Ethnopharmacology, 35: 191-196. Sezik, E., Yesilada, E., Honda, G., Takaishi, Y., Yoshio, T. and Toshihiro, T. (2001). Traditional medicine in Turkey X. Folk medicine in Central Anatolia. Journal of Ethnopharmacology, 75: 95-115. Sezik, E., Yesilada, E., Tabata, M., Honda, G., Takaishi, Y., Fujita, T., Tanaka, T. and Takeda, Y. (1997). Traditional medicine in Turkey VIII. Folk medicine in East Anatolia; Erzurum, Erzincan, Agri, Kars, Igdir provinces. Economic Botany, 51: 195-211.
118
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Shuya, C., Xingguo, C. and Zhide, H. (2003). Identification and determination of ecdysone and phenylpropanoid glucoside and flavonoids in Lamium maculatum by capillary zone electrophoresis. Biomedical Chromatography, 17: 477-482. Simic, M., Kundakovic, T. and Kovacevic, N. (2003). Preliminary assay on the antioxidative activity of Laurus nobilis extracts. Fitoterapia, 74: 613-616. Simoes, C.M.O., Schenkel, E.P., Bauer, L. and Langeloh, A. (1988). Pharmacological investigations on Achyrocline satureioides (Lam.) DC., Compositae. Journal of Ethnopharmacology, 22: 281-293. Smith, J.A., Madden, T., Vijjeswarapu, M. and Newman, R.A. (2001). Inhibition of export of fibroblast growth factor-2 (FGF-2) from the prostate cancer cell lines PC3 and DU145 by Anvirzel and its cardiac glycoside component, oleandrin. Biochemical Pharmacology, 62: 469-472. Sokmen, M., Angelova, M., Krumova, E., Pashova, S., Ivancheva, S., Sokmen, A. and Serkedjieva, J. (2005). In vitro antioxidant activity of polyphe noI extracts with antiviral properties from Geranium sanguineum L. Life Sciences, 76: 2981-2993. Soulimani, R., Younos, C., Jarmouni, S., Bousta, D., Misslin, R. and Mortier, F. (1997). Behavioural effects of Passiflora incarnata L. and its indole alkaloid and flavonoid derivatives and maltol in the mouse. Journal of Ethnopharmacology, 57: 11-20. Stevens, P.F. (1978). "Erica L." In: Flora of Turkey and the Aegean Islands. Vol. 6, ed. P.H. Davis, University Press, Edinburgh. pp. 95-97. Su, B., Zhu, Q. and Jia, Z. (1999). Aquaticol, a novel bis-sesquiterpene from Veronica anagallisaquatica. Tetrahedron Letters 40: 357-358. Siileyman, H., Demirezer, L.b., Kuruiiziim, A., Banoglu, Z.N., Go~er, F., bzbak)r, G. and Gepdiremen, A. (1999). Antiinflammatory effect of the aqueous extract from Rumex patientia L. roots. Journal of Ethnopharmacology, 65: 141-148. Siizge~, S., Meri~li, A.H., Houghton, P.J. and Cubuk~u, B. (2005). Flavonoids of Helichrysum compactum and their antioxidant and antibacterial activity. Fitoterapia, 76: 269-272. Szoke, E., Maday, E., Tyihak, E., Kuzovkina, LN. and Lemberkovics, E. (2004). New Terpenoids in Cultivated and wild chamomile (in vivo and in vitro). Journal of Chromatography B, 800: 231-238. Tabata, M., Sezik, E., Honda, G., Yesilada, E., Fukui, H., Goto, K and Ikeshiro, Y. (1994). Traditional medicine in Turkey III. Folk medicine in East Anatolia, Van and Bitlis provinces. International Journal of Pharmacognosy, 32: 3-12. Tandan, S.K, Chandra, S., Tripathi, H.C. and Lal, J. (1990). Pharmacological actions of seselin, a coumarin from Seseli indicum seeds. Fitoterapia, 61: 360-363. Taniguchi, M., Fujiwara, A., Baba, K and Wang, N.-H. (1998). Two biflavonoids from Daphne acutiloba. Phytochemistry, 49: 863-867. Tepe, B., Sokmen, M., Akpulat, H.A. and Sokmen, A. (2005). In vitro antioxidant activities of the methanol extracts of four HelLchrysum species from Turkey. Food Chemistry, 90: 685-689. Toker, G., Kupeli, E., Memisoglu, M. and Yesilada, E. (2004). Flavonoids with antinociceptive and anti-inflammatory activity from the leaves of Tilw argentea (Linden). Journal of Ethnopharmacology, 95: 393-397. Tokuda, H., Konoshima, T., Kozuka, M. and Kimura, R. (1991). Antitumour activities of triterpene saponins from Verbascum songaricum. Oncology, 48: 77-80. Tsao, R., Yang, R. and Young, J.C. (2003). Antioxidant isoflavones in osage orange, Maclura pomifera (Raf.) Schneid. Journal of Agriculture & Food Chemistry, 51: 6445-6451. Tunalier, Z., Kosar, M., Kupeli, E., Cal}!;, 1. and Baser, KH.C. (2007). Antioxidant, antiinflammatory and antinociceptive activities and composition of Lythrum salicaria L. Extracts. Journal of Ethnopharmacology, 110: 539-547. Tunon, H., Olavsdotter, C. and Bohlin, L. (1995). Evaluation of anti-inflammatory activity of some Swedish medicinal plants. Inhibition of prostaglandin biosynthesis and PAF-induced exocytosis. Journal of Ethnopharmacology 48: 61-76. Turker, A. and Camper, N.D. (2002). Biological activity of common mullein, a medicinal plant. Journal of Ethnopharmacology, 82: 117-125.
Effects of Medicinal Plants m Turkey
119
Tuzlaci, E. and Erol, M.K (1999). Turkish folk medicinal plants. Part II: Egirdir (lsparta). F~toterapia, 70: 593-610. Tuzlaci, E. and Erya~ar Aymaz, P. (2001). Turkish folk medicinal plants, Part IV: Gtmen CBalikesir). Fitoterapia, 72, 323-343. Vasudeva, S.M. (1999). Economic importance of pteridophytes. Indian Fern Journal, 16: 130-152. Vieitez, E. and Ballester, A. (1972). Phenolic and coumaric components in Erica cinerea. Anales del Instituto Botanico A.J. Cavanilles, 29: 129-42. Villar, A., Gasco, M.A. and Alcaraz, M.J. (1984). Anti-inflammatory and anti-ulcer properties ofhypolaetin-8-glucoside, a novel plant flavonoid. Journal ofPharmacy and Pharmacology, 36: 820-823. Weiss, R.F. (1988). Herbal Medicine. Baconsfield, Baconsfield, pp. 313-314. Wicht!, M. and Bisset, N.G. (Eds) (1994). Visci herba-Mist!etoe herb. In: Herbal Drugs and Phyto-pharmaceuticals. CRC Press, Stuttgart, pp. 534-536. Willis, A. (1973). Dictionary of Flowering Plants and Fern, 8th edition, Cambridge University Press, Cambridge, pp. 624-626. Yesilada, E. (1991). Bitkilerde Antienflamatuvar Aktivite Tayininde Kullanl'lan yontemler. (Methods employed for the anti-inflammatory activity assessment in plants). In: IXth Bitkisel Illa~ Hammaddeleri Toplantisi Bildiri Kitabi", (Ed. Ba~er, KH.C.), Eskisehir, Turkey pp. 94-109. Yesilada, E. (2002). Biodiversity in Turkish Folk Medicine. In: "Biodiversity: Biomolecular aspects of biodiversity and innovative utilization". Ed. Bilge Sener. Kluwer Academic! Plenum Publishers, London, pp. 119-135. Yesilada, E., Honda, G., Sezik, E., Tabata, M., Fujita, T., Tanaka, T., Takeda, Y. and Takaishi, Y. (1995). Traditional medicine in Turkey V. Folk medicine in inner Taurus Mountains. Journal of Ethnopharmacology, 46: 133-152. Yesilada, E., Honda, G., Sezik, E., Tabata, M., Goto, K and Ikeshiro, Y. (1993). Traditional medicine in Turkey IV. Folk medicine in the Mediterranean Subdivision. Journal of Ethnopharmacology, 39: 31-38. Yesilada, E. and Kupeli, E. (2002). Berberis crataegcna DC. Root exhibits potent antiinflammatory, analgesic and febrifuge effects in mice and rats. Journal of Ethnopharmacology, 79: 237-248. Yesilada, E. and Kupeli, E. (2007). Clematis vitalba L. aerial parts exhibit potent antiinflammatory, antinociceptive and antipyretic effects. Journal of Ethnopharmacology, 110: 504-515. Yesilada, E., Sezik, E., Honda, G., Takaishi, Y., Takeda, Y. and Tanaka, T. (1999). Traditional medicine in Turkey IX. Folk medicine in north-west Anatolia. Journal of Ethnopharmacology, 64: 195-210. Yesilada, E. and Sezik, E., (2003). Part 28. A Survey on the Traditional Medicine in Turkey: Semi-quantitative Evaluation of the results. In: Recent Progress in Medicinal Plants. Vol.VII. "Ethnomedicine and Pharmacognosy- II" Eds. V.K Singh, J.N. Govil, S. Hashmi, G. Singh. Studium Press, LLC, Houston, Texas, pp. 389-412. Yesilada, E., Tanaka, S., Tabata, M. and Sezik, E. (1989). The anti-inflammatory activity of the fractions from Eryngium billardieri in Mice. Phytotherapy Research, 3: 38-40. Yesilada, E., Tanaka, H., Takaishi, Y., Honda, G., Sezik, E., Momota, H., Ohmoto, Y. and Taki, T. (2001). In vitro inhibitory effects of Daphne oleoides ssp. oleoides on inflammatory cytokines and activity-guided isolation of active constituents. Cytokine, 13: 359-364. Yesilada, E., Ustiin, 0., Sezik, E., Takaishi, Y., Ono, Y. and Honda, G. (1997). Inhibitory effects of Turkish folk remedies on inflammatory cytokines: interleukin-lu, interleukin113 and tumor necrosis factor u. Journal of Ethnopharmacology, 58: 59-73. Zheng, W.F., Shi, F. and Wang, L. (2006), Analgesic activity of ethanol extract from root of Daphne genkwa. Chinese Traditional and Herbal Drugs, 37: 398-402. Zia, A., Siddiqui, B.S., Begum, S., Siddiqui, S. and Suira, A. (1995). Studies on the constituents of the leaves of Nerium oleander on behaviour pattern in mice. Journal of Ethnopharmacology, 49: 33-39.
"This page is Intentionally Left Blank"
5 Comparative Study of the Antigenotoxic Effects of Some Selected Natural Plant Products Against Methylmethane Sulphonate Induced Genotoxic Damage in Cultured Mammalian Cells YASIR HAsAN SIDDIQUE 1,* AND MOHAMMAD AFZAL 1 ABSTRACT In the present study the effect of ascorbic acid (20, 40 and 80 pM), nordihydroguaiaretic acid (NDGA) (O.S, 1.0 and 1.S pM) and apigenin (S, 10 and 20 pM) was studied against the genotoxic damage induced by 60 pM of methylmethane sulphonate on human lymphocytes culture using chromosomal aberrations (CAs) and sister chromatid exchanges (SCEs) as a parameter. The treatments result in a dose dependent decrease in the CAs and SCEs, suggesting a protective role during the chemotherapy of cancer patients, as there are chances of developing secondary tumors. Ascorbic acid was most effective in reducing the genotoxic damage as compared to NDGA and apigenin, and apigenin was more effective as compared to NDGA i.e. Ascorbic acid> Apigenin> NDGA. The results of the present study suggest that ascorbic acid, NDGA and apigenin can modulate the genotoxicity of MMS in human lymphocytes in vitro. Key words : Ascorbic acid, nordihydroguaiaretic acid, apigenin, chromosomal aberrations, sister chromatid exchanges
INTRODUCTION The herbal preparations are traditionally used for the treatment of various diseases and for that reason it becomes necessary to assess the mutagenic 1. Human Genetics and Toxicology Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh - 202 002, U.P., India. * Corresponding author: E-mail:
[email protected]
122
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
potential or modulating action of natural plant products when associated with other substances. Herbal medicine is the root of various traditional medicine systems around the world. About 25% of the crude drugs are directly obtained from the botanicals and another 25% derived from chemically altered natural products. Various traditional medicine systems around the world, including ancient Chinese medical system, Indian medicinal system and Amazonial ethnomedicine, rely heavily on herbs for health preservation (Jain et al., 2008). Despite the advances in pharmacology and synthetic organic chemistry the dependence on natural plant products remain unchanged (Fostar, 1999). Apigenin is one of the several active ingredients found naturally in many fruits and vegetables, including apples and celery. It is found in several popular spices, including basil oregano, tarragon, cilantro and parsley. A high amount of apigenin is found in parsley, peppermint, lemon, berries and fruits (Peterson & Dwyer, 1998). It is a member of flavone family of the flavonoid. Apigenin is recognized in traditional or alternative medicine for its pharmacological activity (Hoftman, 2000). In several strains of Salmonella typhimurium it was found negative for reverse mutation assay (NLM, 2000). It induced SOS repair system in E. coli K-12 stain in PQ37 with and without metabolic activation (Czeczot & Bilbin, 1991). Apigenin has also been reported to have anti-inflammatory effects, free radical scavenger, anticarcinogenic (Kim et al., 1998), grown inhibitor in various cancer lines (Yim et al., 2001; Wang et al., 2000) and antigenotoxic properties (Siddique et al., 2008; Khan et al., 2006). N ordihydroguaiaretic acid (NDGA) is a phenolic lignan, present in the evergreen shrubs, Larrea divaricata and Guaiacam officinale (Agarwal et al., 1991). NDGA possesses a number of interesting biological properties, that are of potential use for humans such as enzyme inhibitor (Capdevilla et al., 1988), antimicrobial (Hurtado et al., 1979), a protector from neurotoxicity and bladder toxicity (Frazier & Kehrer, 1993), stimulator of corpus luteum for the secretion of progesterone (Carlson et al., 1995), potential vas and branchiodialator (Nagano et al., 1996), antimutagenic action against alkylating drugs (Wang et al., 1991), synthetic progestins (Siddique et al., 2006; Siddique et al., 2007) and estrogens (Siddique & Mzal, 2004). Ascorbic acid is a powerful antioxidant, which plays an important role in intra cellular oxidation/reduction systems, and in binding of free radicals produced endogenously (Laurence et al., 1997). Methylmethane sulphonate is a monofunctional alkylating agent with both neoplastic and
Antigenotoxic Effects of Some Selected Natural Plant Products
123
mutagenic activities (Mittal & Musarrat, 1990). It alkylates DNA at the N7 position of guanine and the N-3 position of adenine. All these changes may give rise to abnonnal base pairing at DNA replication (Madrigal-Bujaidar et al., 1998). The present study deals with the comparative study of ascorbic acid, NDGA and apigenin against the genotoxic damage induced by methylmethane sulphonate in cultured human lymphocytes.
MATERIALS AND METHODS Chemicals Apigenin (Sigma), NDGA (Sigma), Ascorbic acid (SRL); PhytohaemagglutininM, antibiotic-antimycotic mixture, RPMI-1640 (Gibco); dimethylsulphoxide, 5-Bromo 2-deoxyuridine, colchicine (SRL, India); Giemsa stain (Merk).
Human Lymphocyte Culture Duplicate peripheral blood culture were perfonned according to Carballo et al. (1993). Briefly, heparinized blood samples (0.5 mL), were obtained from a healthy female donor and were placed in a sterile culture tubes containing 7 mL ofRPMI-1640 medium, supplemented with fetal calf serum (1.5 mL), antibiotic-antimycotic mixture (1.0 mL) and phytohaemagglutinin CO.1 mL). The flasks were kept in an incubator at 37°C for 24 h.
Chromosomal Aberration Analysis After 24 h, 60 ~M of methylmethane sulphonate was given with 20, 40 and 80 ~m of ascorbic acid, 0.5, 1.0 and 1.5 ~M of NDGA and 5, 10 and 20 ~M of apigenin separately and kept for another 48 h. The treatment of selected doses ofMMS, ascorbic acid, NDGA and apigenin were also given separately. After 47 h, 0.2 mL of colchicine (0.2 Ilg/mL) was added to the culture flask. Cells were centrifuged at 1000 rpm for 10 min. The supernatant was removed and 5 mL of prewarmed (37°C) KCI hypotonic solution (0.075 M) was added. Cells were resuspended and incubated at 37°C for 15 min. The supernatant was removed by centrifugation at 1000 rpm for 10 min., and 5 mL of chilled fixative (methanol: glacial acetic acid; 3:1) was added. The fixative was removed by centrifugation and the procedure was repeated twice. The slides were stained in 3% Giemsa solution in phosphate buffer (pH 6.8) for 15 min. 300 metaphases were examined for the occurrence of different type of abnormality. Criteria to classify the different types of aberrations were in accordance with recommendation ofEHC 46 for environmental monitoring of human population OPCS, 1985).
124
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Sister Chromatid Exchanges Analysis For sister chromatid exchange analysis, bromodeoxy uridine (10 ~g/mL) was added at the beginning of the culture. Mter 24 h, 60 ~M of MMS treatment was given with 20, 40 and 80 ~ of ascorbic acid, 0.5, 1.0 and 1.5 ~M of NDGA and 5, 10 and 20 ~M of apigenin separately and kept for another 48 h in an incubator. Mitotic arrest was performed by adding 0.2 mL of colchicine (0.2 ~g/mL). Hypotonic treatment and fixation were performed in the same way as for the chromosomal aberration analysis. The slides were processed according to Perry and Wolff (1970). The sister chromatid exchange average was taken from an analysis of metaphase during second cycle of division.
Statistical Analysis Students t-test was used for the analysis of CAs and SCEs. Regression analysis was also performed for the dose effect of ascorbic acid NDGA and apigenin against the genotoxic dose of MMS using statistical software (Statsoft Inc.).
RESULTS The results of the present study reveal that the treatment of ascorbic acid, NDGA and apigenin reduced the genotoxic effect ofMMS. The used dosage of ascorbic acid, NDGA and apigenin were not genotoxic per se. The treatment of 20, 40 and 80 ~M of ascorbic acid, 0.5, 1.0 and 1.5 ~M of NDGA and 5, 10 and 15 ~M of apigenin with 60 ~ of MMS results in a dose dependent decrease in number of abnormal metaphases (Table 1). The chromatid exchanges and dicentric chromosomes were completely eliminated even at the lowest tested doses of ascorbic acid, nordihydroguaiaretic acid and apigenin (Table 1). In sister chromatid exchange analysis the treatment of 20,40 and 80 ~ of ascorbic acid, 0.5, 1.0 and 1.5 ~M of NDGA and 0.5, 10 and 15 ~M of apigenin with 60 ~M of MMS results in a dose dependent decrease in SCEs/cells (Table 2). Regression analysis was also performed to determine the dose of effect of ascorbic acid, NDGA and apigenin on 60 ~ ofMMS for number of abnormal meta phases and sister chromatid exchanges. A decrease in the slope of linear regression was observed as the dose of ascorbic acid, NDGA and apigenin was increased. For abnormal metaphases the treatment of 20, 40 and 80 ~M of ascorbic acid (F = 3.85; p<0.10; Fig 1), 0.5, 1.0 and 1.5 ~M of NDGA (F = 27.0; p<0.02; Fig 2) ad 5, 10 and 20 ~M of apigenin (F = 7.68; p<0.08; Fig 3) with the increase in the dosage a decrease in the slope of linear regression lines was observed. For sister chromatid exchange analysis, the treatment of 20, 40 and 80 ~M of ascorbic acid (F = 6.57; p<0.063; Fig 4),0.5, 1.0 and 1.5 ~M ofNDGA (F = 30.37; p<0.02; Fig 5) and
Antigenotoxic Effects of Some Selected Natural Plant Products
125
5, 10 and 20 J.1M of apigenin (F =24.14; p<0.024; Fig 6) with the increase in the dosage the decrease in the slope of linear regression lines was observed Table 1. Effects of ascorbic acid, nordihydroguaiaretic acid and apigenin on chromosomal aberrations induced by methylmethane sulphonate Treatments
MMS(IIM) 60 MMS(IJM)+ AA(IJM) 60+ 20 60+40 60+80 MMS(IJM)+ NDGA (IJM) 60 + 0.5 60 + 1.0 60 + 1.5 MMS (IJM) + Apigenin (11M) 60+5 60+ 10 60 + 20 AA(IJM) 20 40 80 NDGA (IJM) 0.5 1.0 1.5 Apigenin (IJM) 5 10 20 Untreated Negative control (DMSO, 51l1/mL)
Chromosomal aberrations
Abnormal metaphases without gaps Number
Mean%:tSE
Gaps
CTB
CSB
CTE
DIC
38
12.67 ± 1.92"
23
22
16
5
4
18 12 10
6.00 ± 1.37b 4.00 ± l.13b 3.33 ± 1.03b
12 8 6
10 8 7
8 4 3
28 24 22
9.33 ± 1.67b 8.00 ± 1.56b 7.33 ± 1.50b
16 14 10
16 14 13
14 12 9
23 16 12
7.67 ± 1.53b 5.33 ± 1.29b 4.00± 1.13b
9 8 5
13 9 7
10 7 5
2 3 4
0.67 ± 0.47 1.00 ± 0.57 1.33 ± 0.66
1 2 2
2 2 2
1 2
4 5 6
1.33 ± 0.66 1.67 ± 0.73 2.00 ± 0.80
2 2 3
2 3 3
2 2 3
2 3 4 2 2
0.67 ± 0.47 1.00 ± 0.57 1.33 ± 0.66 0.67 ± 0.47 0.67 ± 0.47
1 2 2 1 1
2 2 2 2 2
1 2
CTB: Chromatid break; CSB: Chromosome break; CTE: Chromatid exchange; DIC: Dicentric; MMS: Methylmethane sulphonate; AA: Ascorbic acid; NDGA: Nordihydroguaiaretic acid; DMSO: Dimethylsulphoxide; SE: Standard error. aSignificant with respect to untreated (p
126
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Table 2. Effects of ascorbic acid, nordihydroguaiaretic acid and apigenin on chromosomal aberrations induced by methylmethane sulphonate SeEs/cell (mean :t SE)
Treatments MMS(J1.M) 60 MMS (J1M) + AA (J1M) 60 + 20 60 +40 60 + 80 MMS (J1M) + NDGA (J1M) 60 + 0.5 60 + 1.0 60 + 1.5 MMS (J1M) + Apigenin (J1M) 60+ 5 60 + 10 60 + 20 AA (J1.M) 20 40 80 NDGA (J1.M) 0.5 1.0 1.5 Apigenin (J1.M) 5 10 20 Untreated Negative control (DMSO, 5 JllImL)
17.22 ± 0.98a 13.24 ± 0.74b 10.34 ± 0.68b 8.84 ± 0.59b 14.12 ± O.84b 12.28 ± 0.76b 11.32 ± 0.74b 13.68 ± 0.78b 12.02 ± 0.72b 10.42 ± 0.70b 2.14 ± 0.27 2.36 ± 0.32 2.94 ± 0.36 2.56 ± 0.34 3.22 ± 0.40 3.98 ± 0.46 1.72 ± 1.76 ± 1.80 ± 1.52 ± 1.58 ±
0.28 0.30 0.31 0.20 0.26
MMS: Methylmethane sulphonate; AA: Ascorbic acid; NDGA: Nordihydroguaiaretic acid; DMSO: Dimethylsulphoxide; SE: Standard error. aSignificant with respect to untreated (p
9 10
,_~~
20
30
40
50
60
70
60
80
95% confld.
Concentration of apigenin
Fig 1. Effect of ascorbic acid on number of abnormal metaphases induced by 60 J1M of methylmethane sulphonate
127
Antigenotoxic Effects of Some Selected Natural Plant Products Y=30657-6.0OO·X Correlation: r = -.9820
29
it
28
{
27
J9
~ 28
'iii
j
25
~
24
0
.li
§
23
z 22 21 04
"'0..... RegressIon
06
08
1.0
12
14
1.6
95%confid
Concentration of norclihydroguaiarebc acid
Fig 2. Effect of nordihydroguaiaretic acid on number of abnormal metaphases induced by 60 JlM of methylmethane sulphonate Y = 25.000 - .6557 • X Correlation: r = -.9406 24r-----------------------------------~
.,22
! a. 20
i
i " '0
18 16
.li
14
~
12 10 L -____________~___________________"'" 2 10 14 18 22
""-
Regression 95% oonfid.
ConcetratJon of apIgenin
Fig 3. Effect of apigenin on number of abnormal metaphases induced by 60 methylmethane sulphonate
JlM
of
Y = 13.990 - .0682' X Correlation: r = -.9317 14.5,------------------------------,
'8 13.5
1 125
'C
11.5
i~ 10.5 u
i
9.5 8.5 ' -___________________________-"'_----' ""- Regression 10 20 30 40 50 60 70 80 90 95% confid. Concentration of ascorbic acid
Fig 4. Effect of ascorbic acid on sister chromatid exchanges induced by 60 IlM of methylmethane sulphonate
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
128
Y =15.373 - 2.800· X Correlation: r = -.9839 14.6
1'"
14.0
<: .<:
13.4
"0
12.8
e
12.2
• III
.,~
'"~ .<:
0
0
(J
J
11.6 0
"0....
11.0 0.4
0.8
0.6
1.0
1.2
1.6
1.4
Re~reSSlon
95°0 confid.
Concentration of nordihydroguaiaretic acid
Fig 5. Effect ofnordihydroguaiaretic acid on sister chromatid exchanges induced by 60 J.lM of methylmethane sulphonate
=
Y 14.480 - .2091 • X Correlation: r = -.9799
14.0 0
~.,
'"<:
.~
~
13.5 13.0 12.5
"0 III
12.0
e
11.5
'"E
o
ti ~
J!l 11.0
~
10.5
o
10.0 2
6
10
14
18
22
"0.... Regression 95% confid.
Concentration of apigenin
Fig 6. Effect of apigenin on sister chromatid exchanges induced by 60 IlM of methylmethane sulphonate
DISCUSSION The results of the present study reveals that all the natural plant product are effective in reducing the genotoxic damage of methylmethane sulphate. Ascorbic acid is most effective in reducing the genotoxic damage as compared to apigenin and NDGA. In our present study the following trend was found in the potency of reducing the genotoxic damage induced by methylmethane sulphonate-Ascorbic acid> Apigenin> N ordihydroguaiaretic acid. Vitamins act as antioxidants and free radical scavengers, thereby acting as anticarcinogenic, anticlastogenic and antimutagenic agents. Ascorbic acid
Antigenotoxic Effects of Some Selected Natural Plant Products
129
possesses a substantial nucleophilic character and it has been suggested that ascorbate might protect against electrophilic attack or cellular DNA by intercepting reactive agents (Edgar, 1974) or that ascorbyl anion radical with a high extent of unpaired electron delocalization is responsible for the scavenging of free radicals (Bielski, 1961; Szeto et al., 2002). As the treatment with ascorbic acid results in a decrease of the genotoxic damage induced by MMS, it may be due to the scavenging property of ascorbic acid. NDGA possesses antioxidant (Olivetto, 1972) and free radical scavenging property (Nakadate, 1989; Marthy et al., 1990), which may be useful in reducing the genotoxic damage induced by MMS. The doses ofNDGA were selected on the basis of study performed by Madrigal-Bujaidar et al. (1998). Genotoxicity testing provides human a risk assessment. An increase in the frequency of chromosomal aberrations in peripheral blood lymphocytes is associated with an increased overall risk of cancer (Hagmar et al., 1984). Most of the chromosomal aberrations observed in the cells are lethal, but there are many other aberrations that are viable and cause genetic effects, either somatic or inherited. The ready quantifiable nature of sister chromatid exchanges with high sensitivity for revealing toxicant-DNA interaction and the demonstrated ability of genotoxic chemicals to induce significant increase in sister chromatid exchanges in cultured cells has resulted this endpoint being used as indicator of DNA damage in blood lymphocytes of individuals exposed to genotoxic carcinogens (Albertini et al., 2000). The above genotoxic endpoints are well known workers of genotoxicity of any reduction in the frequency of these genotoxic endpoints gives an indication of the antigenotoxicity of a particular compound (Albertini et al., 2000). Genotoxic effect of anti cancer drugs in non-tumor cells are of special significance due to the possibility that they may induces secondary tumors in cancer patients. Therefore, the antigenotoxic effects of these natural plant products are of special significance, that they are potent enough in reducing the genotoxic damage, thereby reducing the possibility of developing secondary tumors in the patients undergoing chemotherapy, but care should be taken with regard to their concentration if consumed in isolation.
ACKNOWLEDGEMENTS Thanks are due to DST, New delhi for the project No: SRIFTILS-003/2007 under SERC Fast Track Scheme for young scientists to the author Yasir Hasan Siddique and to the Chairman, Department of Zoology, AMU, Aligarh-202002 for the laboratory facilities.
REFERENCES Agarwal, R., Wang, Z.Y., Bik, D.P. and Mukhtar, H. (1991). Nordihydroguaiaretic acid, an inhibitor oflipoxygenase, also inhibits cytochrome P-450-mediated monoxygenase activity in rat epidermal and hepatic microsomes. Drug Metabolism Disposition, 19: 620-624. Albertini, R.J., Anderson, D., Douglas, G.R., Hagmar, L., Heminiki, K., Merelo, F., Natrajan, A.T., Norppa, H., Shuker, D.E.G., Tice, R., Walters, M.D. and Aitio, A. (2000). IPeS
130
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
guidelines for the monitoring of genotoxic effects of carcinogens in humans. Mutation Research, 463: 111-172. Bielski, AH.J., Allen, O. and Schwarz, H.A (1981). Mechanism of disproportion action of ascorbic radicals. Journal of American Chemistry, 103: 3516. Capdevilla, J., Gil, L., Orellana, M., Marmett, 1.J., Mason, J.L., Yadagiri, P. and Falck, J.R (1988). Inhibitor of cytochrome P-450 dependent arachidonic acid metabolism. Archives of Biochemistry and Biophysics, 261: 257-263. Carlson, J.C., Sawada, M., Boone, D.L. and Stauffer, J.M. (1995). Stimulation of progesterone secretion in dispersed cells of rat Corpora lutea by antioxidants. Steroids, 60: 272-276. Czeczot, H. and Bilbin, M. (1991). Effect of flavones and their metabolites on induction of SOS repair in the strain PQ37-E.coli K-R Acta Biochimica Polonica, 38: 71-74. Edgar, J.A (1974). Ascorbic acid and biological alkylating agents. Nature, 248: 136-137. Foster, S. (1999). Medicinal plant development in the United States. In: Special Forest Products: Biodiversity meets the market place (General Technical Report GTR-WO-63). Ed. by Vance, N.C. and Thomas, J., Washington, DC: USA Forest Service. Frasier, L. and Kehrer, J.P. (1993). Effect of indomethacin, aspirin, nordihydroguaiaretic acid, and piperonyl butoxide on cyclophosphamide induced bladder damage. Drug Chemical Toxicology, 16: 117-133. Hagmar, L., Bonassi, S., Stromberg, V., Brogger, A., Knudson, J.E., Norppa, H. and Reuterwall, C. (1998). Chromosomal aberrations in human lymphocytes predict human cancer: A report from the European study group on cytogenetic biomarkers and health (ESCH). Cancer Research, 58: 4117-4121. Hoftman, D.L. (2000). Chamomile, Herbal Materia Medica < http://www.inpicentor.com/ librarylherballmaterial-mediallchamomile.asp>. Hurtado, L., Hernandez, R, Hernandez, F. and Fernandez, F. (1979). Fungitoxic compound in the Larrea resin. In: Centro de Investigacion en Qumija Aplicada Ed. by Campos, E., Mabri, T.J. and Fernandez, S., Mexico, pp. 328-340. Jain, A, Porwal, P., Jain, AK. and Bhargava, S. (2003). Arthritis and Herbs, In: Recent Progress in Medicinal Plants, Vol. 19, Ed. by Singh, V.K., Govil, J.N. and Sharma, RK., Studium Press LLC, USA, pp. 91-105. Khan, T.H., Jahangir, T., Prasad, L. and Sarwat, S. (2006). Inhibitory effect of apigenin an benzo (a) pyrene mediated genotoxicity in Swiss albino mice. Journal of Pharmacy and Pharmacology, 58: 1655-1660. Kim, H.P., Mani, 1., Iversen, L. and Ziboh, V.A. (1998). Effect of naturally occurring flavonoids and bioflavonoids on epidermal cycloxygenase and lipoxygenase from guinea pigs. Prosta glandons Leukot Essential Fatty acids, 58: 17-24. Laurence, D.R., Dennett, P.N. and Brown, M.J. (1997). Clinical Pharmacology, eighth Edition, New York, Churchill Living Stone. Madrigal-Bujaidar, E., Diaz Barriga, S., Cassani, M., Marquez, P. and Revuelta, P. (1998). In vivo and in vitro antigenotoxic effect nordihydroguaiaretic acid against SCEs induced by methylmethane sulphonate. Mutation Research, 419: 163-168. Marthy, S.N., Cooney, C.G. and Clearfield, H.R. (1990). Hydrogen peroxide-induced alterations in prostaglandins secretion in the rat colon in vitro. Inflammation, 14: 645-661. Nagano, N., Imaizuni, Y., Hirano, M. and Watanabe, M. (1996). Opening of Ca 2+-dependent K+ channels by nordihydroguaiaretic acid in porcine coronary arterial snoop muscles cell. Japanese Journal of Pharmacology, 70: 281-284. Nakadate, T. (1989). The mechanism of skin tumor promotion caused by phorbal esters: possible involvement of arachidonic acid cascade/lipoxygenase, protein kinase C and calcium/calmodulin systems. Japanese Journal of Pharmacology, 49: 1-9. NLM (2000). CCRIS (Chemical Carcinogenesis Research Information System), National Library of Medicine, Bethesda, MD (Record Nos. 3789, 3790). Olivetta, E.P. (1972). Nordihydroguaiaretic acid. A naturally occurring antioxidant. Chemical Industry, 2: 677-679. Peterson, J. and Dwyer, J. (1998). Flavonoids: dietary occurrence and biochemical activity. Nutrition Research, 18: 1995-2018.
Antigenotoxic Effects of Some Selected Natural Plant Products
131
Rithidech, K.N., Tungjai, M. and Whorton, E.B. (2005). Protective effect of apigenin on radiation induced chromosomal damage in human lymphocytes. Mutation Research, 585: 96-106. Siddique, Y.H. and Afzal, M. (2004). Antigenotoxic effect of nordihydroguaiaretic acid (NDGA) against SCEs induced by estradiol-17~ in human lymphocyte chromosomes in vitro. Indian Biologist, 36: 25-27. Siddique, Y.H., Ara, G., Beg, T. and Afzal, M. (2007). Protective role of nordihydroguaiaretic acid (NDGA) against the genotoxic damage induced by ethynodioldiacetate in human lymphocytes in vitro. Journal of Environmental Biology, 28: 279-282. Siddique, Y.H., Beg, T. and Afzal, M. (2006). Protective effect of nordihydroguaiaretic acid (NDGA) against norgestrel induced genotoxic damage. Toxicology in Vitro, 20: 227-233. Siddique, Y.H., Beg, T. and Afzal, M. (2008). Antigenotoxic effect of apigenin against anticancerous drugs. Toxlcol in Vitro, 22: 625-63l. Snyder, RP. and Gillies, P.J. (2002). Evaluation of the clastogenic, DNA intercalation, and topoisomerase II interactive properties of bioflavonoids in Chinese Hamster V79 cells. Environmental and Molecular Mutagenesis, 40: 266-276. Szeto, Y.T., Tomlinson, D. and Benzie, I.F.F. (2002). Total antioxidant and ascorbic acid content of fresh fruits and vegetables: implications for dietary planning and food preservation. British Journal of NutritIOn, 87: 55-59. Wang, W., Heideman, L., Chung, C.S., Pelling, J.C., Koehler, K.J. and Birt, D.F. (2000). Cell cycle arrest of G21M and growth inhibition by apigenin in human colon carcinoma cells lines. Molecular Carcmogenesis, 50: 102-110. Wang, Z.Y., Agarwal, R., Zhou, Z.C. and Bickers, D.R (1991). Anti-mutagenic and antitumorigenic activities of nordihydroguaiaretic acid. Mutation Research, 261: 155-162. Yin, F., Giuliano, A.E., Law, RE. and Van Herle, A.J. (2001). Apigenin inhibits growth and induces G21M arrest by modulating cyclin-cdk regulators and ERK-MAP kinase activation in breast carcinoma cells. Anti Cancer Research, 21: 413-420.
"This page is Intentionally Left Blank"
6 Inflammation and Mediators S. SINGH 1,*,
P.
A. KOULl, R. SHARMA1, G.D. SINGHl, A. KIiAJuRIAl AND V.K. GUPTAl
KOULl,
ABSTRACT Inflammation is the normal protective response to tissue injury caused by any noxious stimulus to destroy the invading organisms, remove irritants and set stage for tissue repair. It is triggered by the release of different cellular and humoral mediators from the injured tissues and migrating cells. The specific mediators vary with the type of inflammatory process. Enormous numbers of them are known today. It has been said that medical knowledge doubles every 10 years. With this assumption, if we start with one mediator in the early twenties we end up with a very high number till today. The concept of inflammatory mediators may sound reasonably limited yet the field has become so specific that no single individual is known who masters it all. As the number of mediators has grown, so has the number of targets. Unti/1930's the microcirculation attracted all the attention, in late 30's existence of accumulation of leukocytes in terms of mediators i.e., endogenous substances capable of attracting white blood cells out of the blood vessels. 70's was the era of prostaglandins which created revolution in the history of inflammation and ended with COX-1 and COX-2. Today any cell is a fair target for mediators. The functional response may be extremely subtle but a cell can be induced to express or reveal a new antigen on its surface without any visible change or the target cell simply becomes activated. It is well known that all inflammatory cells can exist in these two conditions - resting and activated - and this may ultimately become true for all cells. Activated cells then produce new mediators of their own and the situation become more complicated. An attempt has been made in this review 1. Indian Institute ofIntegrative Medicine, Canal Road, Jammu Tawi - 180001, India. Corresponding author: E-mail:
[email protected]
*
134
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
to explain some of the hidden facts of inflammation through explaining the mediators of inflammation. Key words : Inflammation, mediators, cells, prostaglandins, cytokines, leukotrienes, selectins, integrins INTRODUCTION Ever since the dawn of history, man has always tuned to plants for alleviation of his sufferings. Herbal drugs have been used in the treatment of diseases since time immemorial. Written history on the use of medicinal and aromatic plants can be traced back to about 1000 years. Four fifth of the population in the world lives in the developing countries where majority of the people benefit from traditional medicines and only 15% ofthem benefit from allopathic medicines. Asia is the most important continent for the traditional medicinal plants. India, being an agricultural country, is richly endowed with a wide variety of indigenous floras, some of which have long been employed for their medicinal use. Several indigenous systems of medicines are practiced in India such as Ayurveda, Unani, and Siddha etc. All of them use a variety of herbs, minerals and other biological products as medicaments since long. These natural products are used in traditional medicines on the basis of subtle principles and concepts. These principles are basically similar in all the indigenous systems of Ayurveda, Unani, and Siddha. These are vividly described in Ayurvedic classics as a series oftexts called "nighantus" especially devoted to the usage of herbal drugs and their properties. The recent upsurge of global interest in herbal drugs has created a fresh impetus on the development of plant-based medicines and health products.
As science progresses, the unknown diseases are being discovered and reported. The diseases, which were rarely heard of, are becoming very common. With the advancement in the drug development system, many new drugs are flooding the world market but still they have not succeeded over the drudgery of diseases. Uncounted number of dreadful diseases has been listed in the medical history; inflammation is one among them and a concerned disease particularly rheumatoid arthritis. Although, inflammatory diseases have affiicted a large proportion of world population thus crippling the activity of human beings without killing, yet, no permanent cure. Every individual, a human or an animal must defend itself against multitude of different pathogens including viruses, bacteria, fungi, protozoan, metazoan parasites as well as tumours and a number of various harmful agents which are capable to derange its homeostasis. Inflammation is a complex stereotypical body's reaction to invasion by any infectious agent, antigen challenge or even just physical or traumatic damage.
Inflammation and Mediators
135
To talk of inflammation, war is the perfect metaphor for it. Both are necessary evils, more-or-Iess displaying stereotyped responses to outside threats. The specialized troops (white cells), suicide-commandos (neutrophils), long term siege armies (granulomas), supply routes (vessels), communications and intelligence (mediators), and a huge array of lethal weapons (inflammatory enzymes) come to patrolling the site of inflammation and surmount an inflammatory response. Despite idealistic rhetoric about "the laws of war", when the fighting starts, there is really only one law: ''kill your enemy". Like it or not, if you want peace, you must be prepared to fight under certain conditions. If you want to be healthy, your body must be able to mount an inflammatory response. Force will always rule our world. Our best hope is that this will be the force of good laws. And the best for which we can hope from the inflammatory response is that, for most of our lives, it will do us more good than harm. "Big Robbins" defines "inflammation as the reaction of vascularized living tissue to local injury". Inflammation is the name given to the more or less stereotyped ways our tissues respond to noxious stimuli, with blood vessels and white blood cells as its twin center pieces and a host of proteins as actors. Inflammation destroys, dilutes, or walls off the injurious agents and sets in motion the limited powers of the body to heal it. The main purpose of inflammation is to bring fluid, proteins and cells from the blood into the damaged tissue for the defense. In other words inflammation, a localised protective event, elicited by injury, is a complex and multimediated process. It may be considered as an essential protective and normal process to any noxious stimulus that may threaten the well being of the host ranging from a transient, self limiting and localised to a complex sustained response involving a part or the whole organism that may lead to the immunological rejection of foreign tissue. When tissue injury is caused by a single finite event, the inflammation and reparative process progresses smoothly from injury to healing. In these circumstances, the whole inflammatory process is truly beneficial and provides an example of homeostatic mechanism restoring the affected tissue to its former normal healthy state. In contrast, when the injurious agent is self replicating parasite, the inflammatory response becomes much more complex because some form of tissue injury will continue till the agent persists. Living tissue's early local response to an injury is acute inflammation, whereas, if acute inflammation persists for longer time it turns to chronic inflammation which is often systemic and biochemically different from acute inflammation, meaning thereby that inflammation can be acute or chronic depending upon the injurious agents, although acute and chronic inflammatory events often occur simultaneously. The physiological features of inflammation have been recognised since Celsus described them in first century A.D. Acute inflammation is
136
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
recognised by cardinal signs of redness, oedema, heat, pain and loss of function. In fact acute inflammation is not a disease, but a defensive response to injury; however, it is not always beneficial. Acute inflammation comprises the immediate and early response to an injurious agent. The events in the acute inflammatory process are largely mediated by the production and release of a variety of chemical mediators. Despite the diversity, the injurious influence and the tissue involved in inflammation, similar chemical mediators are released, and hence the acute inflammatory process is virtually stereotyped. Further more, the acute inflammation may remain confined to the site of injury and evoke only local signs and symptoms or may be extensive and induce systemic signs as well as evolve secondary line of defense, such as lymphoid tissue. The protein rich fluid and white cells usually polymorphs, migrate from blood that accumulate in the extravascular spaces as a result of an inflammatory reaction, constitute an exudate indicating that acute inflammatory reactions are exudative, while on the other hand chronic inflammation is largely a proliferative one. Furthermore, inflammatory process has been described in terms of the immediate reaction to injury, which gives rise to acute inflammation. The acute response is characterised principally by the vascular and exudative changes. The white cells that participate in acute inflammation are usually neutrophils and macrophages. In contrast, chronic inflammation results from injurious stimuli that are persistent, often for weeks or months leading to a predominating proliferative rather than an exudative reaction. Chronic inflammatory reactions are also characterized by white cell accumulations, chiefly macrophages and lymphocytes and some times plasma cells. Thus, leucocytic exudate in chronic inflammation is mononuclear, whereas in acute inflammation it is polymorphonuclear. Chronic inflammation may arise following acute inflammation or the response may be chronic almost from the onset. Acute to chronic transition occurs when the acute inflammatory response cannot be resolved, due to either the persistence of the injurious agent or to some interference in the normal process of healing. Chronic inflammation is associated with a range of inflammatory diseases, such as rheumatoid arthritis, hepatitis, chronic pulmonary disease, various inflammatory diseases of gastrointestinal tract and chronic periodontal diseases. Chronic inflammatory diseases show a pathological immune response probably due to the presence of perpetuating antigenic determination. Under certain conditions immune reactions are set up against the individual's own tissue, leading to autoimmune diseases. In these diseases auto antigens evoke a self perpetuating immunological reaction that results in several chronic inflammatory diseases, such as rheumatoid arthritis and chronic thyroiditis.
Inflammation and Mediators
137
Some inflammations are difficult to categorise as acute or chronic because there is no sharp line that divides them. Arbitrarily it is said that when an inflammation lasts more than four to six weeks it is said to be a chronic inflammation. However, much depends on the effectiveness of the host response and nature of the injury. Time limits are without meaning. The differentiation of acute and chronic inflammation can best be made on the basis of morphological pattern of the reaction.
INFLAMMATION It is presumed that inflammation is a salutary process. It may have the injurious effects on the body. Teleologically, it is a process of self defenses against noxious stimulus to a certain limit, beyond that it requires drug therapy to curtail the limits of the process. Broadly inflammation is a complex cellular and biochemical response to injurious stimuli that involves a multitude cell types and mediators. It is classified into acute and chronic types. The account of acute and chronic inflammation often separates the two conditions by a description of healing and regeneration. An understanding of the histological picture of chronic inflammation is certainly assisted by knowledge of the process involved in repair, but the separation from acute inflammation leaves the impression that these two varieties of inflammation are distinct from each other, although sharing the term inflammation in their name. In general, the term acute, as applied to inflammation, refers to a response with a rapid onset and relatively short duration characterized by particular vascular phenomena. The common sequel is resolution of the inflammatory exudate and restoration of normal structure and function. In some cases the acute events lose their intensity, so that the process continues as a smoldering chronic condition. As long as irritation persists, the lesion should be considered inflammatory and once the irritation abates or is removed, the process becomes reparative.
Acute Inflammation The inflammatory process has been described in terms of the immediate reaction to injury which gives rise to acute inflammation. The physiological features of acute inflammation have been recognised since Celsus described them in the first century A.D. as the cardinal signs. These are redness, oedema, pain and heat. The fifth sign i.e., loss of function was added later by Galen (1978). Infact acute inflammation is not a disease, but a defensive response to injury, however, it is not always beneficial. In human beings inflammation occurs in vascular tissues, and is primarily a local vascular response where protein rich fluid and cells are brought to the site of injury to neutralize the damaging agent. The damaging agent can be due to a variety of causes viz., mechanical damage (cuts, blows etc.), chemical damage (acids, alkalies, toxins etc.), radiation, burns, scalds, viruses, bacteria, parasites, insect bites, stings, and local pathology such as antigen-
138
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
antibody reactions. Early events in inflammation are obviously vascular in nature and these can be demonstrated on the skin by stricking it with a ruler. Initially a white line is produced because of mechanical stimulus to local capillaries, which have responded with vasoconstriction when blood is pushed out of the vessels. This is a transitory phenomenon, but if sufficient pressure is applied a red line results which is because of vasodilation where the new dilated capillaries fill with blood and produce the red color. The sharpness of the red line fades in about thirty seconds to one min and the area of pressure takes on a dull red, blotchy appearance. This is known as flare and is caused by arteriolar dilation. If sufficient force was applied in the initial trauma a wheal will develop due to the loss of fluid through the vascular wall, resulting in the formation of oedema. As the oedema developed some of the redness may be lost where pressure from the collected fluid close down the capillaries. Vasodilation, in particular, arteriolar dilation increases the local blood supply which, being at a higher temperature than surface tissue, gives a feeling of warmth. These vascular changes are due to chemical mediators being released at the site of injury. The early changes in the blood vessel wall and the changes in the blood flow are known as "stasis". As the fluid leaves the blood with the result it becomes more viscous and the flow slows down, the erythrocytes tend to stick together in the form of rouleaux and push the white cells towards the vessel walls. The collision of white cells with the vessel endothelium lining stimulates them and they increasingly adhere to the vessel wall and this adherence of cells to the vessel wall is known as pavementing of cell. Changes also occur in the endothelial cells of the vessel wall. A sticky film containing fibrin, which entraps polymorphonuclear leukocytes and other blood cells such as platelets, covers their surface. The passage of fluid through the blood vessel walls is due to changes in the hydrostatic and osmotic pressure. In acute inflammation the arteriolar dilation increases the flow of blood in the inflamed region and the pressure on small blood vessels. Eventually when the pressure in the capillaries exceeds the osmotic pressure of the proteins, fluid will leave the vessels and infiltrate into the tissues and form an exudate. Normal interstitial fluid has a low protein content and low specific gravity. In acute inflammation there are changes in the permeability of blood vessel wall, which allow both proteins and cells to pass through it. Although the passage of plasma proteins through the vessel wall is a passive process but cells leave by a different mechanism. The first cells to leave the blood vessels are the PMNLs, but if inflammation persists, monocytes follow and after some time become the dominant cells infiltrating into the inflamed tissue. Erythrocytes can also leave the blood vessels by a passive movement known as diapedesis.
Inflammation and Mediators
139
Mter they leave blood vessels, white cells show directional movement towards the injured tissue. About 90% of PMNLs are neutrophils. Infact the neutrophils are the typical cells of acute inflammation. In vitro neutrophils move faster than the monocytes but the neturophils are short lived, whilst monocytes and macrophages are comparatively long-lived cells. These factors may explain why neutrophils are predominant in acute inflammation, but monocytes, and macrophages and other cells such as plasma cells are predominant in established or chronic inflammations.
Chronic Inflammation Chronic inflammmation may arise following acute inflammation or the response may be chronic almost from the onset. Chronic inflammation usually occurs when acute inflammation remains unresolved due to either the persistence of the injurious agent or to some interference in the normal healing process. Chronic inflammation is associated with a range of inflammatory diseases such as rheumatoid arthritis, hepatitis, chronic obstructive pulmonary disease, various inflammatory diseases of gastrointestinal tract and chronic inflammatory periodonal disease. Chronic inflammatory disease shows a pathological immune response probably due to the presence of a perpetuating antigenic determination. Cells derived from three lines viz., mononuclear phagocyte, lymphoid cells and fibroblasts infiltrate the tissues. In addition there may be granulocytic leukocytes, particularly eosinophils. Macrophages, epitheloid cells, or multinucleated giant cells represent mononuclear phagocytes. Lymphoid cells are present as lymphocytes, plasma cells, or immunoblasts. Fibroblasts produce collagen and ground substance and proliferate to form more fibroblasts. At varying stages during the course of the lesion, lymphocytes, fibroblasts, or granulocytes may predominate, but the most important unit of chronic inflammation is almost always the macrophage and its derivatives, the epitheloid cells and the giant cells. Necrosis is also common in chronic inflammatory lesions. The overall participation of the dividing cells and their efforts in producing new tissue have resulted in chronic inflammation, being described as proliferative, in contrast to the exudative features of acute inflammation. In acute inflammation, macrophages disappear in a few days because of death of the cells, their removal by lymphocytes or migration elsewhere. In chronic inflammation the persistence of macrophages may result from their local proliferation, from longevity of the cells in the tissue, or from their continuing mobilization from the bone marrow. Although the mechanism of inflammation has been studied extensively, it is only recently that major attention has been paid to the target areas available for the pharmacological attack. Only a few drugs help to cure the disease. Many drugs assist the body's own healing process by holding a disease at bay, thereby allowing the nlltural defenses to build
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
140
up or recover after the initial onslaught of disease. Other drugs such as non-steroidal antiinflammatory drugs merely relieve the symptoms of disease such as the pain and swelling in arthritis, without actually affecting the disease process. The major target areas identified for the pharmacological attack are as followed. 1.
Factors responsible for the onset of inflammation.
2.
Mediators released during initial injury
3.
Non specific endogenous antiflammatory factors evoked by the injured tissue.
4.
The processes attempting to restore normal function.
These target areas are inter-related as a chain reaction and the interruption of this chain at any point helps and lead into the success.
Biochemical Events A series of characteristic changes occur in the small blood vessels and the related tissues within the damaged area, when the living tissues are injured. The reaction in the first few hrs. is more or less independent of the nature of the injurious agent. The biochemical events that form the basis of acute inflammation are triggered by a number of compounds. The biochemical changes occur simultaneously and aid numerous changes. The different compounds with diverse chemical structure and actions aiding to inflammation are called inflammatory mediators, which are derived both from tissues and vascular components of the damaged area. Most of the cells release histamine and eosinophil chemotactic factors from their granules and also form and release prostaglandins, leukotrienes (Brain et al., 1985) and platelet activating factor. Serotonin, a vasodilator in man, does not produce significant change in permeability in rats, however, it appears to be a potent inflammatory agent. (Declerk & Harman, 1983). Neutrophil release LTB 4 , a potent chemotactic agent for stimulating and recruiting more neturophils (Bray et al., 1981) may also release several neutral and acid proteases as well as cationic protein that stimulates the release of the contents of the mast cell granules (Safayhi et al., 1992). The lymphocytes release a variety of peptides called lymphokines that inhibit the migration of macrophages, neutrophils, basophils, and eosinophils, exert cytotoxic actions, induce lymphocyte proliferation or induce vascular permeability. Their peptides include interleukins and interferans (Camp et al., 1986). The kinin system, the complement system and the clotting and fibrinolytic system of plasma provide more mediators than influence inflammation. These mediators enhance the vascular symptoms of inflammation. Stimulation of factor XII or Hageman's factor in the clotting system activates pre-kallikrin to kallikrin, which gets converted into
Inflammation and Mediators
141
kininogen and then to bradykinin, as well as activates more factor XII. The latter converts plasminogen pro activators to plasminogen activator that converts plasminogen to plasmin. This, in turn activates the complement system that mediates a series of peptides and cytotoxic agents.
MEDIATORS OF INFLAMMATION It is recognised that inflammation differs from species to species. In same species from one tissue to another and also in the same tissue according to the type of traumas. Yet the early inflammatory events induced by various trauma in different species and tissues have much in common, as far as sign and symptoms are concerned (Ferreira & Vane, 1979). Several highly active substances are liberated locally in tissue during inflammatory reactions. In different types of inflammation, some mediators may have more prominent role than others. The sequence of mediator release may also be important. For instance, in anaphylactic shock there is an explosive and simultaneous release of histamine, SRS-A, PGE 2 and PGF 2<1 and rabbit aorta contracting substance (Piper & Vane, 1971). However, in the inflammatory response to subcutaneous injection of carrageenan in rat, there is sequential release of the mediators (Willis, 1969) at first; there is an output of histamine followed by the detection of bradykinin. Mter 3 h little PG activity «5 ng/mL) was observed but this concentration gradually rose to an average plateau of 80 ng/mL between 18 h to 24 h.
Once the initial reaction to injury has developed, the subsequent changes within the damaged area depend upon the severity, nature and duration of action of the injurious agents. If this is of brief duration or is rapidly and successfully overcome by the defense mechanism of the host, the inflammatory changes will either resolve completely or subside leaving a variable amount of scar tissue with in the injured area. Some types of persistent stimuli lead to massive continued cell migration and to suppuration (pus formation). Cells and tissue changes caused by mediating substances that originate from various cells or plasma are the salient features of the inflammatory process. Stimuli, which trigger the inflammatory process, activate these mediators or lead to their release. Different groups of cells contain several potent mediators. The cells most studied in this respect are neutrophills (polymorphonuclear neutrophills leukocytes), basophills, mast cells and platelets and to a lesser extent macrophages and lymphocytes. These mediators are released in a sequential manner from the inflammatory site by the effected organelles in which histamine is the first to come followed by serotonin bradikinin, members of prostaglandin series and the other related mediators. The onset of inflammation begins with the migration of various inflammatory cells at the site of inflammation. Once inflammatory cells arrive at the site of inflammation, they release mediators, which control
142
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
the later accumulation and activation of other cells. However, inflammatory reactions initiated by the immune system, the ultimate control are exerted by the antigen itself, in the same way as it controls the immune response. For this reason, the cellular accumulation at the site of chronic infection, or in autoimmune reactions, is quite different from that at sites where the antigenic stimulus is rapidly cleared. Inflammatory mediators are soluble, diffuseable molecules that act locally at the site of tissue damage and infection and more distant sites. These mediators are a) Exogenous mediators b) Endogenous mediators
Exogenous Mediators The bacterial products and toxins can act as exogenous mediator of inflammation. Notable among these is endotoxin or LPS of gram negative bacteria. The immune system of higher animals has probably evolved in a variable sea of endotoxin, so it is not surprising that this substance evokes powerful responses. For example, endotoxin can trigger complement activation, resulting in the form of anaphylatoxins C3a and C5a, which cause vasodilation and increase vascular permeability. Endotoxin also activates "Hageman factor", leading to activation of both the coagulation and fibrinolytic pathways as well as the kinin system. In addition, endotoxin elicits T cell proliferation as have been described as superantigen for T cells.
Endogenous Mediators These mediators are produced from within the immune system itself, as well as other systems. For example they can be derived from molecules that are normally present in the plasma in an inactive form. Mediators of inflammation are also released at the site of injury by a number of cell types. These cell types either contain them as preformed molecules within storage granules, e.g., histamine or rapidly switch on the machinery to synthesize the mediators when required e.g., metabolites of aracidonic acid such as prostaglandins. Further more the inflammatory mediators are early phase mediators and the late phase mediators. The early phase mediators are produced by the mast cells and platelets and are especially important in acute inflammation which includes mainly histamine the first to come at the site of inflammation followed by serotonin and other vasodilators like bradykinin. The chemoattractants like C5a and cytokines such as interleukine-1 (IL-1), IL-6 and tumor nacrotic factor-a are also early phase mediators. Late phase mediators are responsible for the regulation of vascular event occuring later after 6 to 12 h ofthe initiation of inflammation these include the products of arachidonic acid.
Inflammation and Mediators
143
Histamine Histamine is a multifunctional substance exerting influence on many types of cells and physiological processes through a distinct set of G proteincoupled receptors. Generally, histamine modulates inflammation and allergic responses via Hl receptors (Ash & Schild, 1966), gastric acid secretions through H2 receptors (Black et ai., 1972), and neurotransmitter release in the central nervous system via H3 receptors (Arrang et ai., 1983). All of these histamine receptor types are members of the supper family of G protein coupled receptors (Lovenberg et ai., 1999). Recently cloning and characterization of a new class of histamine receptor, H4 has been reported (Nakamura et ai., 2000; Liu et ai., 2001). These receptors clearly documented the expression of H4 receptor mRNA on variety of cell types including peripheral blood mononuclear cells, neutrophils, eosinophils, mast cells and resting CD4+ cells. These findings are highly suggestive of a role for the H4 receptor in immune and lor inflammatory modulation. Histamine is the most important vasoactive inflammatory mediator released at the site of inflammation by the inflammatory cells. It is one of the primary mediators released and plays an important and pivotal role in the development and further progression of inflammatory reaction. However, the contents of histamine dwindle within the first sixty min and antihistaminics have no effect on the delayed permeability response. Thus, histamine is mainly important in the early phase of inflammatory responses and in immediate IgE mediated hypersensitivity reaction. Being a biologically potent low molecular weight endogenous biogenic amine, it is distributed in mast cells and basophil granules. It is also present in human platelets. The release of histamine from platelets (platelet release reaction) is stimulated when platelets aggregate after contact with collagen, thrombin, ADP and antigen- antibody complexes. Platelet aggregation and release are also stimulated by platelet activating factor derived from mast cells during IgE-mediated reaction. It is stored in mast cells and basophils largely complexed to mucopolysaccharide such as heparin. It is synthesized from L-histidine by histidine decarboxylase. Two forms of histamine have been described viz., a storage or bound form in the mast cells and intrinsic or free form. Its wide distribution would appear to make it ideally suited as a mediator of acute inflammation and of acute anaphylactic reaction. Widely distributed in the inflammatory cells, histamine has diverse functions including primary, local dilation of small vessels, widespread arteriolar dilation, local increased vascular permeability by contracting endothelial cells, the contraction of non vascular smooth muscles, chemotaxis for eosinophils and blocking T-lymphocyte function. It acts mainly on the microcirculation through Hl receptors (Buss, 1979). The H2 receptors mediate a number of antiinflammatory effects, including the inhibition of eosinophil chemotaxis but cause the vasodilation.
144
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
It appears, therefore, that the role of histamine in the inflammatory response is transient and other mediators and mechanisms are required to explain the continued vascular and cellular changes characterizing the progression and regression of the overall response.
Serotonin Serotonin (5-hydroxytryptamine) is a biogenic amine that was first discovered in the gut (Vialli & Erspamer, 1933). Serotonin was later shown to correspond to a vasotonic substance found in the serum and was therefore called serotonin (Rapport et ai., 1948) Serotonin is also found in brain (Twarog & Page, 1953) and is involved in a wide range behaviors such as sleep, appetite, inflammation and pain perception, locomotion, thermoregulation and sexual activity (Wilkinson & Dourish, 1991). Furthermore, serotonin drugs are used in the treatment of a number of pathological states such as migraine, depression, and anxiety (Sleight et ai., 1991). The multiple action of serotonin is mediated by the specific interaction of this amine with several receptors. Pharmacological and physiological studies identified distinct receptors that were designated 5HT-1A, 1B, 1e, 1D, 2,3, and 4 (Saudou & Hen, 1994). However, some pharmacological studies suggested the existence of additional serotonin receptor subtypes. Further, the classification of 5HT receptor subtypes takes into account not only their pharmacological profile and coupling with second messenger but also their sequences (Saudou & Hen, 1994). Except for 5HT-3 receptors, which are ligand-gated ion channel receptors, all other 5HT receptors belonging to the large family of receptors that interact with G protein. Serotonin is the most important vasoactive mediator that is stored in the mast cell and basophil granules in rodents. It is an endogenous biogenic amine, which, like histamine, has potent effect on small blood vessels and smooth muscles of certain mammalian species. The possible role of serotonin in acute inflammation has not yet been clearly described. In many animal species, mast cells and platelets contain considerably large amount of serotonin which is released along with histamine by many of the inflammatory agents viz., dextran, turpentine and egg albumin (Singh, 1983). Serotonin increases vascular permeability when injected subcutaneouly in small amounts. Furthermore, in some of the studies it has been observed that animals depleted with serotonin showed poor response to local inflammatory reaction produced by different oedemogenic agents. Serotonin is also capable of dilating capillaries producing contraction of nonvascular smooth muscles. Most of it is stored in the gastrointestinal tract and central nervous system but a large amount is stored in the dense granules of platelets.
Inflammation and Mediators
145
Bradykinin Kinins are produced from precursor kininogen via the action of proteolytic enzymes. Two major biochemical cascades for the production of kinins have been described. One within the cardiovascular system activated in association with the blood clotting and another in many other tissue systems activated following tissue injury and inflammation. Bradykinin is the major kinin liberated in the plasma, while in tissues kallidin (Lysyl-bradykinin) is the major kinin formed. Both bradykinin and lysyl-bradykinin are rapidly degraded by kininases to generate the active metabolite des-Arg9 bradykinin or des-ArglO kallidin, respectively, which also contribute to inflammation and hyperalgesia (Dray & Perkins, 1993). Bradykinin has been more extensively characterized than kallidin. It exerts several proinflammatory effects, including (a) plasma extravasation; (b) stimulation offree radical, prostaglandin, and cytokine production from a variety of cells; (c) stimulation of postganglionic sympathetic neurons to affect blood vessels and to release prostanoids; (d) degranulation of mast cells to release histamine and other proinflammatory mediators; (e) chemotaxis of lymphocytes to the sites of injury; and (D mitogenic activity, possibly relating to tissue repair (Dray & Urban, 1996). In addition bradykinin is one of the most potent endogenous algogenic substance which induces pain by stimulating nociceptors directly (skin, joint, muscle) and by sensitizing them to other stimuli. Indeed there is a strong synergism between the actions of bradykinin and other algogenic substances such as PGs and 5-hydroxytryptamine. The effects of kinins are mediated via two main classes of G proteincoupled receptors, B1 and B2. Additional evidences for B3 receptor have been provided by antagonistic studies through pharmacological and molecular techniques (Farmer, 1995). In humans, B1 and B2 receptors are composed of 353 and 364 amino acids respectively representing distinct gene products. Thus couple primarily through the Gq/ll family ofGTP-binding proteins (Hall, 1997). The B2 receptor is expressed more commonly in many tissues and is the preferred site of action for bradykinin and kallidin. Studies with selective antagonists have shown that B2 receptors make a major contribution to inflammatory pain and hyperalgesia (Sterauka et al., 1988; Heapy et al., 1993; Perkins et al., 1993). On the other hand, B1 receptor is encountered less frequently but can be constitutively expressed in certain tissues such as blood vessels. This receptor is preferentially activated by the metabolites des-Arg 9 bradykinin or des-Arg lO kallidin. B1 receptor expression markedly increases during inflammation or infection. The significance of this behavior is not entirely clear but under these circumstances, B1 receptor makes an important contribution to the hyperalgesia (Dray & Perkins, 1993).
146
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Several potent B2 antagonists with analgesic and antiinflammatory activity have been made. These include the peptide analogues viz., NPC567, Hoe 140, NPC16731, the dimeric peptide CP-0127 (bradycor) and a non peptide antagonist, WIN 64338 (Hock et al., 1991; Cheronis et al., 1992; Sawutz et al., 1994). Less is known about the B1 receptor, but it has little homogeneity with the B2 receptor (Mauke et al., 1994), and in keeping with current pharmacological studies, antagonists for this receptor will likely be different from those developed at B2 sites. However, both B1 and B2 receptor antagonists are likely to be useful analgesic and antiinflammatory agents. The kinins, bradykinin and lysylbradykinin, are important mediators of inflammatory response. In asthma also they mediate inflammation with potent bronchoconstrictor actions. The endogenous release of nitric oxide may inhibit bradykinin-induced bronchoconstriction (Kharitonov et al., 1999). They are liberated from precursor molecules, kininogens, by the action of various proteases, collectively known as kininogenases. Three types ofkininogens have been identified viz., high- and low molecular weight kininogen and T-kininogen. These molecules are synthesized by the hepatocytes and are released into the plasma, where in addition to releasing kinins, they function as cofactor in the cogulation pathway, inhibitors of cysteine protease enzymes and part of the acute phase response. Three different pathways may lead to kinin formation during inflammation i.e., the generation of bradykinin as a result of activation of the Hageman factor and the production of plasma kallikrein, the production of lysylbradykinin by tissue kallikreins and the action of cellular proteases in kinin formation. As bradykinin is such a potent peptide, its activity and its formation must be carefully controlled. Activation of the pathway is controlled internally by the presence of inhibitors for each ofthe active components. Complement component C1 inhibitor controls the activity of the activated Hageman factor, while a2- macroglobulin and C1 inhibitor act as kallikrein inhibitor. There are a variety of enzymes in plasma that control bradykinin activity, including carboxypeptidase N, which removes the C-terminal arginine residue, thus inactivating the peptide.
Prostaglandins Prostaglandins belong to a family of chemical messengers, which have been studied extensively over the last 20 to 25 years. Prostaglandins were first identified in semen, and received their name because at that time they were thought to origin from the prostate gland. However, it is now well known that the prostaglandins in semen are synthesized mainly by the seminal vesicles, and the prostate makes a small contribution. Further, it has been discovered that prostaglandins are produced in almost all tissues of the body, and are ubiquitous messenger molecules with a variety of actions. They are fatty acids synthesized from arachidonic acid, a long chain
Inflammation and Mediators
147
polyunsaturated fatty acids, and have been classified into groups from prostaglandin A to prostaglandin I according to structural variation in the five-carbon ring at one end of the molecule. There is further subdivision based on the number of double bonds in the two side chains. Prostaglandins are key mediators of inflammation and play critical physiological role in tissue homeostasis and function. Tissue injury and certain other stimuli such as inflammatory stimuli cause the local release of chemical mediators such as prostaglandins (Bolten, 1998). During an inflammatory reaction, there is a rapid activation and recruitment of inflammatory cells such as macrophages and neutrophils, release of numerous cytokines and enzymes, and production of pro-inflammatory mediators. Prostaglandins are synthesized from arachidonic acid, the initial step of which is catalyzed by the enzyme cyclooxygenase. They are implicated in the inflammatory response and in sensitizing nociceptors to the actions of other mediators. Occuring during the acute and chronic inflammatory illness, prostaglandins are produced at the site of inflammation where it is believed that they mediate many ofthe symptoms of inflammation, such as oedema and pain. Arachidonic acid, precursor of prostaglandins, is released from cell membranes by phospholipases. Cyclooxygenase catalyzes the addition of molecular oxygen to arachidonic acid to form initially the endoperoxide intermediate prostaglandin G2 (Peskar & Maricic, 1998). The same enzyme also possesses peroxidase activity that catalyzes the reduction of this prostaglandin to form PGH 2 • PGH 2 may then react with a number of enzymes sometimes called isomerase to become one of the prostagladins or thromboxanes. The prostagladins are characterized by a five-membered ring which determines the type of prostaglandin. Two products of this reaction are PGD 2 and thromboxane A2. Two other prostaglandins are produced, viz., PGE 2 and PGI 2 , which act as mediators of the vascular phases of inflammation. They are both potent vasodilators. In addition, they act synergistically with certain of the vasoactive mediators, such as histamine and kinnis, to increase vascular permeability. They also stimulate osteoclastic bone resorption suggesting that bone erosion in chronic inflammatory disease may be mediated at least in part by prostaglandins. Prostaglandins are found throughout the body in almost all tissues. The physiological actions of prostaglandins are medited through an interaction with specific receptors coupled to different secondary messenger system that ultimately produce an array of biologic effects. The same pathway used to generate prostaglandins for the inflammatory process is used to produce prostagladins in other tissues. Generally, the prostaglandin produced in a tissue is a function of specific cell type. For example, endothelial cells produce primarily the vasodilatory PGI 2 , platelets generate
148
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
the vasoconstrictor ~, renal prostaglandins are involved in regulation of renal blood flow and prostaglandins in the gastric mucosa have cytoprotective or, more appropriately termed, gastroprotective functions. The major difference in the production of prostaglandins is the specific type of cyclooxygenase used in the pathway. Until recently, it was believed that there is only one type of cyclooxygenase throughout the body. However, Simons and colleagues (1989) identified a second form of cyclooxygenase, COX-2 distinguishing it from the originally identified enzyme (Bjorkman, 1998b). Subsequent work has shown that the two enzymes have a similar size, nearly identical enzyme kinetic, and have 75% amino acid homology, with complete preservation of the catalytic sites for cyclooxygenase and peroxidase activity. The genes for the two enzymes are found on different chromosomes. The two enzymes are known as cyclooxygenase-l (COX-I) and cyclooxygenase-2 (COX-2). Under normal homeostatic conditions, COX-l is a constitutive enzyme -present in most cells and tissues-where it functions to produce physiologically important prostaglandins. COX-l and COX-2 enzymes differ significantly both in protein structure and mRNA. The human cDNA for COX-2 has been cloned and expressed (Hla & Neilson, 1992). It encodes a 70 kD polypeptide of 604 amino acids that is 61% identical to human COX-I. The mRNA of COX-2 consists of 4.1 kilobases and is considerabely larger than the 2.8 kilobase mRNA of COX-I. This is consistent with its inducible nature. Cells use COX-l to produce prostaglandins in the continuous response to stimulation by circulating hormones and other factors (DeWitt, 1995). This differs significantly from the role of COX-2 and enzyme that is rapidly induced at the site of inflammation and is responsible for the production of pro-inflammatory prostaglandins (Masferrer et al., 1996; Peskar & Maricic, 1998). Under normal conditions, resting neutrophils, macrophages, endothelial cells, and fibroblasts show little or no COX activity but in response to an inflammatory stimulus there is increased activity due to synthesis of COX-2 (Richardson & Emcry, 1995) distribution studies indicate the presence of COX-l in nearly all normal issue including kidney, spleen, stomach, liver, heart and brain. In contrast virtually no expression of COX-2 in normal tissues is detectable. COX-2 expression is readily induced in response to proinflammatory stimuli in cells in vitro and in vivo, including macrophages, monocytes, synovial cells and endothelial cells. COX-2 is not expressed to a significant degree in resting tissue, except in brain, in reproductive organs and kidneys (DeWitt, 1995). The function or activity of COX-2 in these tissues is not known yet. In the gastrointestinal tract, prostaglandins PGE-2 and PGI-2 are derived from COX-l activity and have been correlated to the long term protection of mucosal lining from damage (Frolich, 1997). COX-l is found in the blood platelets where it is essential for the synthesis ofthrombaxane
Inflammation and Mediators
149
A-2. COX-1 activation leads to the production of prostacyclin which is ant-ithrombogenic when released from the endothelium and cytoprotective when released from the gastric mucosa. This cytoprotection of the gastric mucosa extends to both exogenous damaging substances as well as endogenously produced gastric juice. In the kidney, prostaglandins are synthesized mainly in the renal medula, the ascending loop of Henley, and the cortex. Renal prostaglandins influence several functions including, total renal blood flow, distribution of renal blood flow, sodium and water reabsorbtion and renin released. These functions with the possible exception of renin secretion seem to depend on COX-1 activity. Renin secretion has been linked to COX-2 activity because the presence of COX-2 has been demonstrated in the macula dens a and found to up-regulate with sodium deprivation. A close link between the inflammatory response and COX-2 induction is becoming evident (Frolich, 1997). On the basis of studies using proinfalmmatory and anti-inflammatory cytokines to induce inflammation, COX2 is induced in macrophages, while the anti-infalmmatory cytokine, IL-10 down-regulates COX-2 expression. Synovia obtained from patients suffering from osteoartritis and rheumatoid arthritis shows COX-2 in mononuclear cells, endothelial cells of blood vessels and synovial fibroblast cells. The evidence for the involvement of prostaglandins in arthritic diseases is extensive (Williams & Higgs, 1988; Goodwin, 1991; Malmberg & Yaksh, 1992). The classical inflammation of rheumatoid arthritis consists of redness, oedema, pain and heat. Proataglandins are found at the sites of inflammation and prostaglandins 1-2, E1, E2 and D2 cause vasodilation leading to redness (Robinson, 1991). Although prostaglandins 1-2, E2, and F2 are only weakly active in producing oedema, they contribute synergestically to the tissue swelling induced by other inflammatory mediators such as bradykinin and histamine. The role of prostaglandins in fever is complex and involves interlukin-1 (Rothwell, 1992). The discovery by Vane in 1971 that NSAIDs such as aspirin exert their antiinflammatory activity by inhibiting the COX enzyme has found wide acceptance although part of the mechanism of action of these compounds probably does not involve prostaglandins (Abramson & Wissmann, 1989; Brooks & Day, 1991). The effect of prostaglandins on the immune system is complex, incompletely understood and often controversial. Among the many actions of prostaglandins on the immune system, inhibition of Band T cell proliferation (Bray, 1987), inhibition of rheumatoid factor (Alvarellos et al., 1988) and suppression of antigen presentation by macrophages, probably are the most relevant effects as far as arthritic diseases are concerned. Although it is generally accepted that prostaglandin E2 is largely immunosuppressive by virtue of its downregulation ofB and T cell functions. It has been recently found that this compound also potentiates Ig class switching both indirectly by inhibiting interferon secretion from TH1 cells
150
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
and directly by enhancing IL-4 induced class switching to IgG and IgE (Phipps et al., 1991). T cells, in particular CD4+ helper cells, playa central role in orchestrating the immune responses that underlie rheumatoid inflammation (Cush & Kavanaugh, 1995). Of the various subsets of helper T cells, the Th1 subset in particular is relevant (Miossec & van den Berg, 1997). Growing attention has been paid to the balance of Th1JTh2 subsets of helper cells. The Th1 subset of cells which functions primarily in cell-mediated immune reaction, seems to play the dominant role in rheumatoid arthritis (Miossec & van den Berg, 1997). Therefore, alteration of the Thlfrh2 balance has been suggested to be a practically valuable therapeutic approach. Prostaglandin E2 has been implicated in the angiogenesis required for the spread of the pannus in the synovial hyperplasia component of rheumatoid arthritis (Colville-Nash & Scott, 1992).
Cytokines During the course of non-specific as well as immunologically mediated inflammatory reactions, many hormone-like mediators called cytokines are produced. They regulate local as well as systemic inflammatory host diffense. Unlike many hormones, cytokines are not normally present in plasma and nor are produced by glands, but are autocrine and paracrine mediators that are produced by inflammatory cells and many other tissues in response to invasive organisms, irritants or trauma. Although monocytes and lymphocytes are the major source of cytokines among blood cells yet evident that cytokines are generated by many other cell types (Cassatella, 1995 & 1999). The ability of polymorphonuclear leukocytes (PMNs) to produce such a variety of cytokines readily suggests that these cells might not only be inflammatory and immune responsives, but also other processes such as hematopoiesis, wound healing, angeogenesis, and antiviral defense. PMNs can express proinflammatory cytokines such as TNFa, (Djeu et al., 1990), IL-1a and IL-1P (Lord et al., 1991) and IL-12 (Cassone et al., 1997), antiinflammatory cytokines such as IL-1ra (Fava et al., 1991). Many cytokines have been identified those contribute either directly or indirectly to inflammatory processes. Cytokines along with some growth factors produce specific protein at the site of inflammation (Kujubu et al., 1991). In general the cytokines are soluble (glyco) proteins, nonimmunoglobulin in nature, released by living cells of the host, which act nonenzymetically in picomolar to nanomolar concentrations through specific receptors to regulate host cell function. They make up the fourth major class of soluble intercellular signaling molecules, alongside of neurotransmitters, endocrine hormones, and autacoids. They possess typical hormonal activities i.e., they are secreted by a single cell type, react specifically with other cell types (target cells) and regulate specific vital functions that are controlled by feedback mechanism. They generally act at short range in a paracrine or autocrine (rather than endocrine) manner.
Inflammation and Mediators
151
They interact first with high affinity cell surface receptors (distinct for each type or even sub type) and then regulate the transcription of a number of cellular genes by little understood second signals. This altered transcription, which can be an enhancement or inhibtion, results in changes in the cell behavior. Target cells, on which cytokines transform their information signal, may be localised in any body compartment (sometimes a long distance from the sight of secretion). Other type ofthese molecules mostly on neighbouring cell in the microenviornment where they have been released. These are characterised as local hormones and their secretion is brought about by autocrine or paracrine mechanisms. During the paracrine secretion some cytokines may escape cell binding and may spill over into general circulation via lymph or plasma. This is important, especially for the products of lymphoid cells which are mobile after having picked up the message in the microenviornment through out the body. Therefore, their immunoregulatory products, (Lymphokines, monokines, interleukins and other cytokines), despite being of local hormone character, may infact act systemically. Cytokines are synthesised, stored and transported by various cell types (Cassatella, 1995) not only inside the immune system (lymphokines, monokines, tumour necrotic factor, and interferons) but also by other which are mainly studied by haematology (colony stimulating factors), oncology (transforming growth factors), and cell biology (peptide growth factors, heat shock and other stress protiens). The central role of cytokines is to control the direction, amplitude, and duration of immune responses and to control the modelling and remodelling of the tissues, be it developmentally programmed, constitutive or unscheduled. Unscheduled remodelling accompanies inflammation, infection, wounding and repair. Individual cytokines can have pleiotropic, overlapping and sometimes contradictory functions depending on their concentration, the cell type they are acting on, and the presence of other cytokines and mediators thus the information which an individual cytokine conveys depend on the pattern of regulators to which a cell is exposed, and not on one single cytokine. It is supported that all cytokines form the specific system or net work of communication signals between cells of the immune system, and between the immune system and other organs. In this inter cell signalling network, the signal is usually transferred by means of specific set of cytokines. Because of the potential and profound biological effects of cytokines, it is not surprising that their activities are tightly regulated, most notably at the level of secretion and receptor expression. Additional regulatory mechanisms are provided by the concomitant action of different cytokines and the presence in the biological fluids of specific inhibitory proteins, soluble cytokine binding factors and specific autoantibodies. The cytokine is a very potent force in homeostasis when activation of the net work is local and cytokines act vicinally in surface-bound or diffusible form, but
152
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
when cytokine production is sustained/or systemic, there is no doubt that they contribute to the signs, symptoms and pathology of inflammation, infections, autoimmune, and malignant diseases. TNFa is an excellent example of such duel action. Locally it has important regulatory and anti tumour activity but when it circulates in higher concentrations beyond the organ of origin, it may be involved in the pathogenesis of endotoxic shock, cachexia and other serious diseases. From the inflammatory point of view there are two main groups of cytokines viz., pro-inflammatory and antiinflammatory. Pro-inflammatory cytokines are produced predominantly by activated macrophages and are involved in the up-regulation of inflammatory reaction (Beauerle & Baltimore, 1996) whereas, antiinflammatory cytokines belong to the T cellderived cytokines and are involved in the down-regulation of inflammatory reactions. The cytokines which play the central role in inflammation are IL-1 and TNFa (Arend & Dayer 1990), because the administration oftheir antagonists, such as IL-Ira (IL-1 receptor antagonist), soluble fragment of IL-1 receptor, or monoclonal antibodies of TN Fa and soluble TNF receptor, all blocks various acute and chronic responses in animal models of inflammatory diseases (Dawson et ai., 1991). Some of these antagonists are beginning to utilize as antiinflammatory agents in diseases such as sepsis and rheumatoid arthritis. IL-1 and TNFa together with IL-6 serve as endogenous pyrogens. The up-regulation of inflammatory reaction is also performed by IL-ll, IFNP, IFNy, and especially by the members of cytokines superfamily. On the other hand, antiinflammatory cytokines (IL4, IL-10 and IL-13) are responsible for the down-regulation of inflammatory response. They are able to suppress the production of pro-inflammatory cytokines. Their strong antiinflammatory activity suggests possible utilization in management of many antiinflammatory diseases, including sepsis, rheumatoid arthritis, inflammatory bowl disease, psoriasis, T cellmediated autoimmune diseases such as type-I diabeties, as well as in acute graft-versus-host disease. IL-10 is capable of effectively protecting mice from endotoxin-induced shock, a lethal inflammatory reaction mediated by TNFa and IL-I (Firstein & Zvaifler, 1997). The production of most of the lymphokines and monokines such as IL-I, IL-6 and TNFa is also inhibited by transforming growth factor (TGFa), but on the other hand, TGFa has a number of pro-inflammatory activities including chemoattractant effects on neutrophils, T lymphocytes, and unactivated monocytes. TGFa has been demonstrated to have in vivo immunosuppressive and antiinflammatory effects as well as proinflammatory and selected immunostimulatory activities. When administered systemically, TGFa acts as an inhibitor, but if given locally can promote inflammation. Generally, TGFa stimulates neurovasculariztation and the proliferation and activities of connective tissue cells and is a pivotal factor in scar formation and wound healing. But TGFa has anti proliferative effect on most other cell types including
Inflammation and Mediators
153
epithelial cells, endothelial cells smooth muscle cells, fetal hepatocytes and myeloid, erythroid, and lymphoid cells. TGFa is a potent immunosuppressive cytokine that suppresses cell mediated as well as humoral immunity including tumor immunity.
Products of the Complement System Complement is a complex system containing more than thirty various glycoproteins present in serum in the form of components, factors, or other regulators and/or on the surface of different cells in the form of receptors. These are present in blood serum in an inactive form and are activated by immune complexes (the classical pathway), by carbohydrates (the lectin pathway), or by other substances, mainly of bacterial origin (the alternate pathway). The components of the classical pathway are numbered 1 to 9 and prefixed by the letter C, e.g., CI, C2 up to C9. CI is composed of three subcomponents CIq, Clr and CIs. The early components of alternate pathway are known as factors and each molecule is named by a letter, e.g., factor B, D, P. The lectin pathway is the same as the classical pathway, only CIq is omitted. All these pathways use in the later stages of activation the same terminal components C5-C9 that form membrane attack complex (MAC). C3 also participates in all pathways. The complement system influences the activity of numerous cells, tissues and physiological mechanisms ofthe body. These effects may involve either the whole complement, or only individual components of fragments. Activation of the complement cascade, with the formation of the effector MAC unit, results in cytotoxic and cytolytic reactions. Target cells for MAC action may be heterologus erythrocytes, nucleated cells, bacteria (gramnegative, susceptible to serum), microscopic fungi, viruses with a surface envelope and viruse-infected cells. The results of the cytotoxic complement reaction may be beneficial for the body (elimination of the infectious agent or damaged cells) or harmful (damage to autologus normal cells by immunopathological reactions). The complement system is a potent mechanism for the initiation and amplification of inflammation. This is mediated through fragments of complement components, which belongs to anaphylatoxins. Anaphylatoxins are proteolytic products of the serine proteases of the complement system C3a, C4a, and C5a. The production of anaphylatoxins follows not only from complete activation, but also from activation of other systems which may directly cleave C3, C4 and C5. Such enzymes include plasmin, kallikrine, tissue and leukocyte lysosomal enzymes, and bacterial proteases. C5a is extremely potent for stimulating neutrophils chemotaxis, adherence, respiratory burst generation and degranulation. C5a also stimulates neutrophils and endothelial cells to express more adhesion molecules. Ligation of the neutrophil C5a receptor is followed by mobilization of the membrane arachidonic acid which is mobilized to prostaglandins and
154
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
leukotrienes including LTB 4 , another potent chemoattractant for neutrophils and monocytes. Following ligation of C5a receptor, IL-l is released. Thus, the local synthesis of C5a at the site of inflammation has powerful pro-inflammatory properties. C3b and C4b fragments act as opsonins and enhance phagocytosis. In addition to inducing phagocytosis, ligation of complement receptor on neutrophils, monocytes and macrophages may also stimulate exocytosis of granules containing powerful proteolytic enzymes and free radical production through the respiratory burst which further act as pro-inflammatory. The activation of complement by immune complexes is normally beneficial. Immune complexes bearing C3b are efficiently removed from tissues and from the circulation by monocytes and other phagocytes. However, there are circumstances in which immune complex production continues at a high level; complement complexes may then prove deleterious. Such complexes may form in tissues, e.g., in glomeruli of patients with autoantibodies to glomerular basement membrane (Goodpasture's syndrome) or at motor end-plates in-patients with autoantibodies to acetylcholine receptors (Myasthenia gravis).
Chemotactic Factors The term chemotaxis refers to the movement of leukocytes (or cells in general), induced by a chemotactic stimulus. Beside chemotaxis, which is the stimulated directional migration, leukocytes also possess two other types of movements viz., random migration which is undirected spontaneous migration and chemokinesis i.e., stimulated unidirectional migration. A chemotactic stimulus is provided by substances that can either attract or repulse the cells. Thus, chemotactic cell movements can be either negative or positive, i.e., the cells may move towards the source of the chemotactic substance (towards an increasing concentration gradient) or in the opposite direction. The positive movement is typical for leukocytes. Substances possessing chemotactic activity are called chemotactic factors (chemotaxins, chemoattractants). Leukocyte chemotaxis is mainly responsible for their mobilization at the inflammatory site. Both exogenous and endogenous chemoattractants participate in this event. Exogenous chemotaxins include bacterial oligopeptides, lectins, denatured proteins, some lipids and lipopolysaccharides. Endogenous chemotaxis are produced by the host organism and are of humoral or cellular type. The humoral endogenous chemotaxins are complement fragment C5a, C5desArg, and Ba, fibrinopeptides, kallikrein and plasminogen activators whereas, the cellular component comprised of LTB 4 , platelet activating factor, and chemotactic cytokines. Interaction between the chemotactic factor and its corresponding receptor triggers a series of coordinated biochemical events, which include changes in the cell transmembrane potential, altered cyclic neucleotide
Inflammation and Mediators
155
levels and ion flow across the cytoplasmic membrane and increased glucose utilization and oxygen consumption. The composition of membrane phospholipids is altered and arachidonic acid, released by phospholipases, is metabolized into a number of biologically active intermediates and products. Within a few min, the leukocyte changes from round to a triangular shape that is oriented along the direction of the chemotactic gradient. Reorganisation of cytoskeletal contractile elements, particularly active microfilaments and microtubular structures, contribute to this shape change. Activation of the contractile cell system not only results in the migration but also in other form of movement such as enhanced adherence, spreading, endocytosis and secretion of lysosomal enzymes resulting in the further stimulation of the inflammatory mediators.
The Acute Phase Reactants Within the spectrum of systemic reaction to inflammation two physiological responses in particular are regarded as being associated with acute inflammation. The first involves the alteration of the temperature set-point in the hypothalamus and the generation of the febrile response. The second involves alteration in metabolism and gene regulation in liver. Three cytokines that are released from the site of injury viz., IL-l, TNFa and IL6 are considered to regulate the febrile response, possibly as a protective mechanism. It is important to consider the acute phase response (and inflammation) as a dynamic homeostatic process that involves all of the major systems of the body, in addition to the immune, cardiovascular and central nervous system. Normally, the acute phase response lasts only a few days; however, in case of chronic recurring inflammation, an aberrant continuation of some aspects the acute phase response may contribute to the underlying tissue damage that accompanies the disease, and may also lead to further complications. The second important aspect of the acute phase response is the radically altered biosynthetic profile of liver. Under normal circumstances, the liver synthesizes a characteristic range of plasma proteins at steady state concentrations. Many of these proteins have important functions and higher plasma levels of these acute phase reactants or acute phase proteins are required during the acute phase response following an inflammatory stimulus. Although most of the acute phase reactants are synthesized by hepatocytes, some are produced by the other cell types, including monocytes, endothelial cells, fibroblasts and adipocytes. Acute phase reactants have a wide range of activity that contribute to host defense i.e., they can directly neutralize inflammatory agents, help to immunize the extent oflocal tissue damage, as well as participate in tissue repair and regeneration. There is a rapid increase in the plasma concentration of many complement cascade components the activation of which ultimately results in the local accumulation of neutrophils, macrophages and plasma proteins. These
156
Camp. Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
participate in the killing of infectious agents, the clearance of foreign and host cellular debris, and the repair of damaged tissue. Coagulation components, such as fibrinogen, play an essential role in promoting wound healing. Proteinase inhibitors neutralize the lysosomal proteases released following the infiltration of neutrophils and macrophages, thus controlling the activity of the proinflammatory enzyme cascade. The major acute phase reactants in mammals include serum amyloid-A and C-reactive protein or serum amyloid-P component depending on the species. Generally, only one of these proteins is an acute phase reactant in a given mammalian species. In humans normal plasma serum amyloid P component levels remain constant during inflammation but C-reactive proteins can increase upto lOOO-folds depending on the disease and its severity. Serum amyloid-A enhances the binding HDL3 to macrophages during inflammation, concomitant with a decrease in the binding capacity ofHDL3 to hepatocytes. The exquisite responsiveness of C-reactive protein to acute phase stimuli, along with its wide concentration range and ease of measurement, have led to plasma C-reactive levels being used to monitor accurately the severity of inflammation and the efficacy ofthe disease management during an infection. Conversely, some diseases are associated with relatively low plasma levels of C-reactive proteins. C-reactive protein and serum amyloidP component are archetypal examples of plasma proteins that are likely to be beneficial during the transient acute phase response but these have detrimental effects in chronic inflammation. Acute phase reactant synthesis is under control performed by inflammatory mediators from which several cytokines and hormones specifically regulate the transcription of human acute phase reactants these include TNFa, IL-l, IL-6, IL-ll, IFNy, TGFa and glucocorticoids. In addition, insulin and akadaic acid have been shown to act as inhibitor of the cytokine-derived induction of some acute phase reactants. Most of the increase in acute phase reactant biosynthesis is due to increase (or decrease) gene transcription.
Caspases Caspases are a family of intracellular cysteine proteases that clear their specific substrates at aspartic acid residues. These proteases have been implicated in two aspects of cytokine biology viz., proteolytic processing and bioactivation of the proforms of certain interleukins such as pro-IL-l~ and pro lL-18 and signaling by apoptosis inducing members of the tumor necrosis factor receptor family, such as tumor necrosis factor receptor-l (TNFR-l) (CD120a) and Fas (CD95). Abundant evidence, derived from molecular biological, genetic, and pharmacological studies, has demostrated a critical role for specific caspases in the bioactivation or bioactivity of particular cytokines. Moreover, these caspases to play nonredundant roles in a wide variety of cytokine mediated inflammatory and autoimmune
Inflammation and Mediators
157
diseases. The involvement of caspases in cytokine biology is examined, with emphasis on relevance to infectious diseases, inflammation and autoimmunity. Ten members of the caspase family of cysteine proteases in humans have been described (Alnemri, 1997; Salvesen & Dixit, 1997). These proteins are initially synthesized as single polypeptide zymogens that undergo proteolytic processing at specific aspartic acid residues to produce the active enzyme. The active, processed caspases are composed of heterotetramers, containing two large and two small subunits, which form two active sites per molecule. Caspases represent a unique family of intracellular cysteine proteases, with absolute specificity for aspartic acid in the P-1 position of substrates. This fact, together with the observation that procaspase zymogens are processed at aspartic acids to produce the active proteases, creates opportunities both for auto activation of caspases and for cascades of caspase activation in which one active member cleaves and activates another. The N-terminal region of the zymogens is typically but not always removed from the protein during processing. Caspases can be grouped into two broad categories (a) the caspase-1 like family and (b) the caspase-3 like family. The categorization of caspases into subfamilies is based on differences in their substrate preference, comparisons of their primary amino acids sequences, and depending on whether their primary function is most relevant to cytokine processing versus apoptosis regulation. The human caspase-1 subfamily consists of caspase-1, 4 and caspase-5. The murine caspase-II protein is probably the orthologue or a close homologue of human caspase-4 (Wang et az', 199B). These caspases share extensive aminoacid sequence identity and are intimately in proinflammatory cytokine processing. The human caspase-3 subfamily consists of caspase-2, caspase-3, caspase-6, caspase-B caspase-9 and caspase-10. Compared to the caspases 1 subfamily, these caspases generally share greater primary amino acid sequence. The members of the caspase-3 subfamily are initimately involved in apoptosis, functioning either as upstream initiators or down stream effectors of this cell death process. Based mostly on differences in their prodomains, the caspase-3 subfamily can be further divided into three additional groups. Abundant evidence implicates caspases in inflammatory and autoimmune conditions mediated by cytokines. The production of bioactive interleukin" a critical mediator of endotoxin shock, is absolutely dependent on caspases-1 in vivo. Similarly, proteolytic processing of prointerleukin-1B, a major inducer of interferon-y in vivo, appears to be mediated exclusively by caspase-l. Cytolytic T-cells and natural killer cells also induce caspase-1 activation. The central importance of caspase-1 as a mediator of inflammatory responses is further supported by the observation that some types of bacteria produce proteins that directly bind to and activate caspase-1, whereas some viruses express proteins that directly inhibit this protease.
158
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Two forms of interleukin-1, IL-1a and 1L-1~ bind to the same receptor(s) explaining their essentially superimpossible bioactivity profiles (Dinarello, 1994). Both IL-1a and 1L-1~ are produced initially as intracellular proproteins which require proteolytic processing to result in secretion from cells. These cytokines are principally produced by activated macrophages, though some other cell types can also produce 1L-1~, including microglial cells, endothelial cells, vascular smooth muscle cells and epidermal langerhans cells. 1L-1~ stimulates proinflammatory responses in neutrophils, endothelial cells, synovial cells, osteoclasts and other cell types (Dinarello & Wolff, 1993). 1L-1 has been implicated in a wide variety of inflammatory conditions in vivo. Multiple animal model experiments have demonstrated a critical role of 1L-1 for inflammatory conditions including arthritis, inflammatory bowel disease. Clinical studies in patients with rheumatoid arthritis suggest that soluble 1L-1R or 1L-1RA may reduce joint tenderness and inflammatory symptoms. (Campion et al., 1996; Drevlow et al.,1996). The in vivo importance of caspase-1 for inflammatory diseases has also been confirmed by animal model experiments using peptide inhibitors of caspase-1 also significantly reduced collagen-induced arthritis in mice. Caspase-1 inhibitors are superior to indomethacin and prednisolne in this animal model of arthritis (Ku et al., 1996). Similarly, the caspase-1 antagonist benzyloxy-valingy alanyl-3(s)-3-amino-4-oxo-5-(2,6-dichlorobenzoyl-oxy acid) ethyll ester, which inhibits LPS - induced IL-1~ secretion from a monocyte cell line with an IC 50 of24 mm, suppressed carrageenan-induced paw oedema in rats by upto 60% when administered 1 h after the inflammatory agent (Elford et al., 1995). Though overexpression of caspase-1 can induce apoptosis (Miura et al., 1993), lack of caspase-1 does not interfere with the normal cell turnover required for tissue homeostasis, caspase-1 knockout mice develop normally and have no apparent increased incidence of tumors or other anomalies (Kuida et al., 1995). Moreover, absence of caspase-1 does not interfere with any immune system functions. The T-cell proliferative responses to listeria monocytogenes and delayed type hypersensitivity reactions for example, are reportedly normal in caspase deficient mice, suggesting that the inflammatory cytokines generated by caspase-1 dependent processing may be more relevant to macrophage-mediated inflammatory responses than to T cell-dependent immune reactions (Gu et al., 1997) Tumor necrosis factor-a (TNF-a) has been implicated at some level in most inflammatory and auto immune diseases, as well as in graft rejection, heart failure and other conditions LPS-induced production ofTNFa is some what reduced in caspase-1 and caspase-II knockout mice (Wang et al., 1998) but probably not enough to be of physiological relevance. Interferon-y, which is linked to the pathogenesis of inflammatory and other autoimmune diseases are elevated indirectly by caspase-l. The indirect mechanism of caspase-1 production of interferon-y has been demonstrated by experiments
Inflammation and Mediators
159
in caspase-1 knockout mice (Gu el al., 1997). Hence caspase-1 represents an attractive target for treatment of autoimmune, inflammatory and other diseases when these cytokines are implicated. It is quite evident that caspase are absolutely required for the production of several proinflammatory cytokines including IL-1a, IL-1~, IL-18 and probably interferon-yo These cysteine proteases also playa requisite role in the signaling mechanisms. Human diseases involving these cytokines therefore, are potentially amenable to modulation by caspase inhibitors. Further investigations of the effects of caspase inhibitors are now required. From this analysis, clinical indications for caspase- inhibiting drugs can be deducted and appropriate clinical trials initiated to test their efficacy in humans.
Nitric Oxide Nitric oxide (NO), a small hydrophobic inorganic gas molecule, highly versatile, potent, short lived and free radical which acts as a unique neuronal messenger molecule for both intra and intercellular messages involved in the regulation of diverse physiological processes including smooth muscle contractility, platelet reactivity, central and peripheral neurotransmission and the cytotoxic actions of immune cells. It is a relatively stable unchanged radical that readily crosses lipid membranes and interacts with a few specific targets. It also reacts with other species possessing unpaired electrons, to yield secondary products that are often more reactive. It has become a species of extreme biological interest due to the diversity of its physiological functions and general ubiquity. NO is crucial for many physiological functions, an inappropriate release of this mediator has been linked to a number of pathologies. Thus agents that modulate the activity of NO may be of considerable therapeutic value, in particular that reduce the formation of NO. It may be beneficial in pathophysiological states in which excessive production of NO is the contributory factor. These include diseases, such as septic shock, neurodegenerative diseases and inflammation. In fact NO is a colorless gas at room temperature and pressure. Its maximum solubility in water is similar to that of molecular oxygen, 2-3 nM. It is a fairly nonpolar molecule which could be expected to freely diffuse through membranes. One of the most unique and outstanding chemical features of NO is that it is a paramagnetic species. Using the most basic bonding description, it is immediately evident that NO has an unpaired electron. Although much of the chemistry of NO is dominated by the fact that it is a radical, it does not possess the type of reactivity normally associated with other radicals. For example, unlike other, carbon, oxygen or nitrogen centered radicals, NO does not even have the tendency to dimerize i.e., at standard temperature and pressure, NO tends to remain in monomeric form. This lack of dimerization has been attributed to the fact
160
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
that overall bonding does not increase when two NO molecules interacted (Palmer et al., 1988) thus, the formal bond order in NO is 2.5, while the bond order for ONNO is 5. 0= N-N=O Bond order 2.5
Bond order = 5 (still 2.5 bonds per NO)
NO which is one of the smallest biologically active messenger molecules has a wide range of physiological and pathological actions. It is synthesized from the terminal guanidino nitrogen atom of L-arginine by the action of cytosolic enzyme nitric oxide synthase (Hibbs et al., 1987, Knowles et al., 1989, and Bredt et al., 1991). Nitric oxide synthases (NOS) constitute a family with atleast three distinct isoforms. Inorder of their molecular characterisation these include neuronal (cNOS) (Murphy et al., 1993), inducible (iNOS) (Leone et al., 1991) and endothelial (ecNOS) (Marietta, 1991) synthases. This nomenclature is based on the tissue of origin from which original protein and cDNA were first isolated. Although the three isoforms have different molecular weights and variable cofactor requirements, all of them are dependent on NADPH, and show similarities with cytochrome P-450 reductase and also with the bacterial enzymes sulfite reductase and cytochrome P-450 BM3. The formation of NO by NOS is linked to incorporation of molecular oxygen into the molecule (Bredt & Snyder, 1990). NOS has been proposed to form NO and L-citrallin in two steps, the first step being the formation of N-hydroxy-L arginine and in the second, its three electron oxidation. Both steps may utilize different heme-based oxidants i.e., a perferryl species, [FeOO]+, for the first step and a peroxoiron species (FeO)3+ for the second step. Both of these are produced when heme reacts with molecular oxygen (Moncada et al., 1991). All forms of NOS contain four prosthetic groups, flavin-adenine dinucleotide, flavin mononucleotide, tetrahydrobiopterin and a heme complex, iron protoporphyrin IX. All NOS isoforms are dependent on calmodulin. In the inducible isoform calmodulin is already present in a tightly bounded form. The role of calmodulin is to control the electron transfer from NADPH to the flavins, possibly by causing a reorientation of the reductase and oxygenase domains of NOS, perhaps moving them into a favourable position for electron transfer between them. The function of tetrahydrobiopterin is unclear, but may substitute for peroxy-heme to carry out nucleophilic attack on hydroxyarginine or perhaps may act as a stabilizer of the active structure of NOS (Moncada et al., 1991). cNOS and ecNOS are the constitutive isozymes and are calcium! calmodulin dependent, whereas iNOS is absent from the mammalian cells but under physiological conditions is induced by proinflammatory stimuli,
Inflammation and Mediators
161
such as bacterial lipopolysaccharide or the cytokines viz., tumor necrosis factor (TNF), interleukin-1 (IL-1), or interferon-y UFN-y), as well as their combination. cNOS and ecNOS constitutive isozymes, when activated, release short "Puffs" of NO from neurons and endothelial cells which is important in both intercellular and intracellular signalling (Charles et ai., 1993). iNOS, the inducible isozyme is induced in many cell types including vascular smooth muscle cells, endothielial cells, hepatocytes macrophages, neutrophils, chondrocytes and cynoviocyted (Stadler et ai., 1991; StefanovicRacic et ai., 1993) in response to inflammatory and immunologic simuli. iNOS generates much larger quantities of NO over longer period of time (nanomoles, rather than picomoles). The NO generated by macrophages activated by cytokine and endotoxin contributed to their cytotoxic and cytostatic properties against target cells (Palmer et ai., 1993). Although NO generated by constitutive NOS appears to be beneficial in many physiological processes, the excess of NO generated by iNOS has been implicated in the pathogenesis of various inflammatory and immunologically mediated diseases including graft-vs-host, diabetes, viral infections and arthritis (Ialenti et ai., 1993) During inflammation profound physiological changes are often observed including vasodilation, fever and activation of the immune system. These changes are induced by a myriad of cellular mediators, such as endotoxin, various cytokines and eicosanoids. It has been established that NO plays an important role as one of these mediators in addition to it's described functions in cell-cell communications and neurotransmission. As the product of three isoforms of NO synthases, it is the iNOS which is expressed by a wide variety of cell types in response to stimulation (Nissler & Billiar, 1993) producing much large quantities of NO relative to the two other constitutive isoforms during sepsis and inflammation. Cell type that expresses iNOS in response to stimulation by certain cytokines and microbial products includes hepatocytes, macrophages, Kupffer's cells and chandrocytes. The regulation of iNOS expression is quite complex, as it involves a variety of mechanisms within a wide range of cell type. For example, the factors that have been important in the rodent for regulating iNOS expression within hepatocytes, as compared to nonhepatocytes, include transcriptional, posttranscriptional as well as posttranslational mechanisms. Regulation of iNOS gene expression is helped by calmodulin molecules to be fully activated at basal levels of calcium allowing the transcription of enzyme. Thus, transcription with subsequent translation of the iNOS gene appears to play an important role in its regulation. Transcription of the iNOS gene is controlled, both positively and negatively, by a number of inflammatory mediators present during infection and inflammation such as interferon- y, tumor necrosis factor ex interleukin-1~, interleukin-2 and lipopolysaccharide (Xie et ai., 1992; Feldman et ai., 1993; Lorsbach et ai., 1993; Vodovotz et ai., 1993). The
162
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
expression of iNOS response to these agents varies among different cell types and there is often strong synergy among many of these agents. The specific mechanisms by which transcription of the iNOS gene is initiated appear to be quite complex and remain to be elucidated, however, the possible regulatory sequences within the promoter, regulatory region in both mouse and human iNOS has been identified. iNOS gene expression can also be regulated at posttranscriptional level, possibly by altering the stability of the newly transcribed mRNA. As L-arginine is the sole physiological nitrogen donor for NO formation, it is clear that regulation of its availability could present one method of controlling cellular rates of NO systhesis. Intracellular arginine may be increased via one of three mechanisms, through uptake from the extracellular fluid, by intracellular protein degeneration, or by endogenous synthesis. There is no evidence that the availability of arginine as a substrate for NO formation is affected by change in the rate of cellular protein degradation. Hepatocytes, macrophages, and pulmonary artery endothelial cells have been shown to up-regulate arginine transport in response to inflammatory mediators that stimulate the production of NO (Bogle et al., 1992; Inove et al., 1993; Lind et al., 1993). NADPH is an essential cofactor in NO synthesis, as it functions as an electron donor (Stuehr & Griffith, 1992). One and a half NADPH molecules are required for each molecule of NO production. Correlation between the formation of NO and the activity ofNADPH-generating pentose phosphate pathway, as well as the rate limiting enzyme glucose-6phosphate dehydrogenase has been made (Blachier et al., 1991; Corraliza et al., 1993). These correlations suggest that NADPH may be up-regulated by the same inflammatory mediators that induce NO formation. Tetrahydrobiopterin, another essential cofactor in NO synthesis (Stuehr & Griffith, 1992) is thought to be involved in the binding of two inactive iNOS isomers into the active dimeric form. During sepsis and inflammation a wide array of cell types are induced to express iNOS, which then produces large amounts of NO, that may then interact with various target molecules, such as oxygen, that groups and metals within the prosthetic groups of various enzymes resulting in their activation or inactivation. In addition, NO may interact directly with DNA as well as with toxic oxygen radicals, either potentiating their toxicity or neutralizing them. The cellular response to NO are dependent on the cell types producing NO as well as the local environment, as the effects of NO appear to be primarily limited to paracrine and autocrine activities as a result of the short biological halflife of NO and its interaction with oxyhemoglobin in the blood stream. Thus, it results in the production of inactive nitrate (N0 3-) and methemoglobin. However, there have been suggestions that NO may exert some endocrine like activity by binding to albumin and glutathione, there by forming stable nitrosothiols that may
Inflammation and Mediators
163
circulate and release active NO at distant sites (Gaston et al., 1993; Keaney et al., 1993). There is evidence that NO can potentiate as well as ameliorate the toxicity of circulating oxygen radicals often seen during sepsis and inflammation as well as during the periods of reperfusion. Rubbo et al. (1995) implicated NO involvement in oxygen radical mediated membrane lipid peroxidation. In contrast, in a perfusion liver model exposed to endotoxin, the inhibition of NO synthesis increases superoxide (0- 2 ) mediated injury (Bautista & Spitzer, 1994). It appears that NO can combine with 0- 2 to form the intermediate peroxynitrite, which is then protonated to peroxynitrous acid. Depending on the pH of the local environment, peroxynitrous acid degrades to either inactive metabolites or toxic radicals. Within an alkaline environment peroxynitrous acid adopts a cis configuration, with subsequent decay to the inactive metabolite nitrate (N0 3 ). However, under acidic conditions peroxynitrous acid adopts trans configuration and yields toxic products, hydroxy radical (OH) and nitrogen dioxide (N0 2 ). Thus, it appears that NO may behave either as a toxic free radical or as an oxygen radical scavenger, depending on the local conditions. The roles of constitutive and inducible NOS isoforms in inflammation have been extensively studied (lalenti et al., 1993). It is likely that NO from constitutive eNOS plays a role in the early stages of inflammation as a mechanism to decrease and limit the process by inhibiting white cell activity and platelet aggregation (Moncada et al., 1990) and by inducing vasodilation (Kajekar et al., 1995). In contrast, NO from iNOS contributes too many aspects of chronic inflammation. In models of acute and chronic inflammation, it has been shown that constitutive NOS accounts for majority of NO activity in early parts of the process, at this point, some polymorphonuclear cells and a few resident macrophages express iNOS. At the peak of chronic inflammations, there is an eightfold increase in total NOS activity, of which greater than 90% can be attributed to iNOS in activated macrophages. The NOS activity is substantially reduced after 14 days as the inflammation disappears. The use of selective iNOS inhibitors may be of benefit in management of chronic inflammatory processes. Chondrocytes in culture express iNOS (Charles et al., 1993) and decreases their proteoglycan synthesis when stimulated by inflammatory cytokines. Adjuvant arthritis in rats in exacerbated by L-arginine and reduced by NOS inhibitors (lalenti et al., 1992). In rheumatoid arthritis elevated concentrations of nitrite and nitrate have been found in synovial fluid of patients, which suggests an increased local release of NO (Farrell et al., 1992). However, it is likely that NO plays a role in down-regulating the activity of osteoclasts. Furthermore, NOS inhibitors have been shown to potentate bone resorption and enhance bone loss in animals.
164
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Psoriasis lesions which is a chronic inflammatory disease, contains high levels of iNOS, which is absent from healthy skin and there is increased expression of iNOS (Bruch et al., 1996). Further inflammatory conditions in which excessive NO production by iNOS has been demonstrated include asthma and inflammatory bowel disease (Allcan & Kubes, 1996). Furthermore, a consequence of the release of early mediators in inflammation is the modulation of threshold and sensitivity of nociceptors leading to inflammatory pain. Prostaglandins play a major role in this process. NO may also be involved but it is not clear at this stage whether NO enhances or decreases the pain threshold. The ability of NO to reduce inflammatory pain may also underlie the anti-nociceptive effects of endogenous compounds such as opiates (Ferreira et al., 1991). Thus, NO may itself possess anti-nociceptive actions. In human studies involving pain in the hand vein, NO also appears to exert nociceptive effects. Following administration of bradykinin or hyperisomolar solution to isolated perfused hand vein, NOS inhibitors markedly reduced the discomfort (Kindgen & Arndet, 1996). As such, this profile of activity suggests a possible therapeutic role for cNOS inhibitors in reducing inflammatory pain.
Free Radicals The role offree radicals have been implicated in causation of several diseases such as liver cirrhosis, atherosclerosis, cancer, diabetes, ageing and inflammatory disorders etc. and the drugs that can scavenge oxygen radical have great potential in ameliorating these disease processes. Although, the body has several mechanisms to protect the action of oxygen radicals but at times these protective mechanisms are not sufficient when compared to the insult caused to the body. Supplementation of non-toxic antioxidants may have a chemoprotective role in the body in these conditions. The superoxide anion (0-), H 2 0 2 and hydroxyl radical (OR-) are the major ROS which induce cell degeneration by increasing lipid peroxidation of cell membrane, break DNA strand and denaturate cellular proteins resulting in further tissue damage. The natural cellular antioxidant enzymes include superoxide dismutase (SOD), which scavenges superoxide radicals by speeding up their dismutation, Catalase (CAT) a haeme enzyme which removes H 20 2 and glutathione peroxidases (GPX), selenium- containing enzyme which scavenges H 2 0 2 and other peroxidases. Detoxification of the superoxide anion is not a terminating step in free radical scavenging, since the enzymes catalysed dis mutation results in the production ofH20 2 which accumulates in the mitochondria and cytosol. Unless the peroxide is scavenged by CAT and GPX, it in the presence of iron, may also lead to production of OR-. These ROS together with singlet molecular oxygen may attack lipids, proteins and DNA of cells following increased lipid peroxidation chain reactions resulting in wide spread cellular injury and ultimately pathophysiological conditions.
Inflammation and Mediators
165
A significant understanding ofthe reactive oxygen intermediates (ROn and reactive nitrogen intermediates (RNI) in inflammatory diseases is underway. During last two decades interest in the role of free radicals in diseases with an inflammatory component, such as rheumatoid arthritis, inflammatory bowel disease, pulmonary emphysema, atherosclerosis, and neurodegenerative disorders, grew rapidly. Most research into the role of free radicals in inflammation was directed toward the putative role of ROI as damaging agent. Many studies centered on the oxidative modification of biomolecules such as 0.1- antitrypsin, cartilage proteoglycans and collagen, low-density lipoprotein, DNA, lipids, immunoglobulin G, and so on (Halliwell & Gutteridge, 1985; Merry et al., 1989). The cytotoxicity of free radicals and their reaction products was also investigated, particularly in relation to enthothelial cell function (Kus et al., 1995) The free radicals were studied in context of a direct, destructive role in biology. In the course of the present decade researchers have increasingly recognized this cellular control aspect of free radical action. Endotheliumderived relaxing factor has been identified as nitric oxide (NO) (Snyder, 1995) and ROI can activate apoptosis, programmed form of cell death, at least under some circumstances (Jacobson, 1996). In these cases, ROI activity plays a critical role in regulating the expression of genes that encode either proteins with potential proinflammatory action, or proteins that may act in a protective fashion such as heat shock protein 32, also known as heme oxygenase (Willis et al., 1996). Fibroblast proliferation and collagen gene expression, associated with the late wound-healing phase of inflammation, have also been linked to intracellular ROI activity (Houglum et al., 1994). Perhaps the key to future antioxidant drug development for the treatment of inflammatory diseases lies in our ability to modulate the previously mentioned ROI-mediated control pathways when they become perturbed as part ofthe inflammatory process. The inappropriate activation of the responses referred to earlier, such as overexpression of the particular genes, excess NO production, and apoptosis, may be involved in disease progression. Nevertheless, it should be remembered that these processes, when properly controlled, are physiologicaL Therefore, ROIIRNI can no longer be regarded solely as damaging species whose complete elimination by antioxidants is bound to have beneficial effects on human health. Indeed, it can be argued that the converse may be the case. In case of treatment of inflammatory diseases, it may be necessary to define the stage of the disease at which a particular antioxidant therapy may most effectively be used, e.g., during acute inflammatory episodes, or in chronic inflammation, or prophylaxis. Another factor that must be taken into account is the individual variation in sensitivity within the population
166
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
(Harris et al., 1994), as well as the different susceptibilities to oxidative stress of different organs (Fraga et al., 1990), tissues (Kristal et al., 1994) and subcellular locations (Richter et al., 1988). An increased susceptibility to the actions of ROI on target lymphocytes of patients with autoimmune disorders could thus be an important mechanism in the pathogenesis of disease such as rheumatoid arthritis by inducing excessive inflammatory responses and increased clonal proliferation of autoreactive lymphocytes via the activation of transcription factors like NF-KB. These considerations pose a challenge in the search for appropriate antioxidant therapies in human inflammatory diseases. It is suggested that the successful therapeutic manipulation of cellular responses by antioxidant drugs will necessitate the maintenance of a critical balance between free radical activity and antioxidant selectively to specific cell types, or even defined subcellular locations, and within a narrow "concentration window" that blocks "inappropriate" cellular responses, but leaves the physiological levels of free radical activity necessary for normal cell function.
Adhesion Molecule Leukocytes are the principal actors in the body's defense system against invading microorganisms (Wissman & Cooper, 1993). This defense system has a nonspecific branch consisting of granulocytes and macrophages and a specific branch of lymphocytes. Granulocytes (neutrophils, eosinophils and basophils) release cytotoxic compounds from their intercellular granules to their local environment when they encounter microorganisms. This random destruction happens rapidly but it may also harm healthy tissue of the body. Macrophages, the other class of defense cells from the nonspecific immune system, can ingest and destroy microorganisms by phagocytosis or, in a similar way to granulocytes by the secretion of cytotoxic compounds. However, macrophages can also act more specifically by collaborating with lymphocytes and their products. The lymphoid system comprises the cellular components responsible for antigen specific immune defense. B-Iymphocytes produce antibodies that bind to foreign organisms and facilitate their destruction, either by activating the complement system which in turn can perforate the membrane, or by opsonizing the microorganisms, i.e., trigger phagocytosis due to receptors for antibodies and the macrophage surface. T-Iymphocytes act mainly by cell-to-cell contact. One subpopulation of T-Iymphocytes recognizes and kills cells which bear foreign antigen; the second subpopulation helps activity of other hemopoietic cells in the immune response or helps to multiply effector cells. All of these leukocytes patrol the body by circulating through the blood and lymphatic system ensuring a continuous surveillance which is a prerequisite for efficient defense (Anderson et al., 1982). Upon tissue damage
Inflammation and Mediators
167
and inflammation, leukocytes are recruited from the blood to sites of injury, and this trafficking displays exquisite specificity (Butcher, 1991; Dunon et at., 1993; Springer, 1994). For instance, neutrophils selectively enter sites of acute inflammation or tissue damage, while eosinophils extravasate into sites of allergic reactions and parasitic infestations. The migration of lymphocytes is even more selective and includes a complex pattern of recirculation that relates to differentiation and activation. To efficiently protect the body from infectious organisms, the cells of the immune system circulate as nonadherent cells in the blood and lymph, and migrate as adherent cells into tissues when necessary. Rapid transition between adherent and nonadherent states is the key to the dual function of immune surveillance and responsiveness. Circulating lymphocytes in the blood have first to adhere to and then to cross the endothelial lining in order to enter the various lymphoid tissues which are involved in recirculation. One exception is the spleen where there is unhampered access of blood leukocytes to parenchyma (Fawcell, 1994). Three classes of adhesion molecules are involved in leukocytes endothelial interactions. These are the selectins, integrins, and members of immunoglobulin (Ig like) superfamily.
Selectins The selectins are a family of adhesive receptors found on leukocytes (L), platelets and endothelial cells (P) or endothelial cells alone (E) (McEver, 1994). They are designated as L-selectin (CD 62L), P-selectin (CD-62P) and E-selectin (CD 62E). They mediate the initial, low-affinity adherence of leukocytes to endothelium, manifested by rolling along the endothelial cell surface under conditions of flow. These receptors belong to a family because they share a common mosaic structure consisting of an aminoterminal C-type lecting domain, a single epidermal growth factor like domain several short consensus repeats similar to those found in regulatory proteins that bind complement, a transmembrane domain, and a short C-terminal cyto plasmic domain. Selectins are known for their binding to carbohydrate ligands via the lectin domain (Rosen & Bertozzi, 1994). L-selectin was first identified as a homing receptor for lymphocyte migration to peripheral lymph nodes (Watson & Bradlley, 1998) and was found to mediate the binding of lymphocytes to high endothelial venules in peripheral lymph nodes. It has since been found to be constitutively expressed on most leukocytes, including lymphocytes, monocytes, neutrophils (PMN) and eosinophils. Leukocytes can be induced to shed Lselectin from their surface by a variety of activating agents such as formylpeptide, phorbol ester, and lipopolysaccharide. P-selectin is stored in the Weibel-Palade bodies of endothelial cells and the a granules of platelets (Hartwell et at., 1998). Its expression on endothelial cells can be induced by a variety of stimuli, including thrombin, histamine, and phorbol
168
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
esters. In response to this stimulation, P-selectin is rapidly redistributed to the cell surface, resulting in expression within min. In some endothelial cells P-selectin is also induced by cytokines or LPS. E-selectin is expressed on the surface of endothelial cells only following stimulation. Tumor necrosis factor-a (TNF-a), interleukin-l (lL-l), Substance P, LPS, and several other stimuli have been found to induce the expression of E-selectin (Vestweber & Blank, 1999). Eselectin expression requires de novo protein synthesis with transcription and translation resulting in peak surface expression in 4-6 h following stimulation in vitro with a gradual decrease to basal levels by 24 h.
Integrins The second class of adhesion molecules involved in leukocyte traffic is the integrin family involved in extracellular material/cell and cell to cell interactions (Hynes, 1992). The term integrin refers to the ability of these molecules to integrate the intracellular media with the extracellular environment. Currently, integrins are considered as a wide family of celladhesion receptors, with a broad pattern of cellular expression that exerts a key role as modulator of major cellular events, such as proliferation differentiation and death. Integrins are transmembrane heterodimers consisting of non-covalently linked a and ~ chains. Integrins are subclassified based on the specific ~ sub unit. The sub classes that have been found to be involved in the leukocyte endothelial cell interactions are the ~2 integrins, the ~l-integrin, and the ~7-integrin. The three ~2 integrins are leukocyte restricted and are composed of a common ~2 sub-unit (CD18) that is noncovalently linked to one of three a subunits. Cell surface expression of ~2 integrins is not sufficient for adhesion of leukocytes to endothelial cells. Activation of the ~2-integrins due to a change in conformation or postreceptor events is required before adhesion occurs (Aplin et al., 1998). This activation is induced by soluble agents like cytokines, chemotactic factors and coagulation factors etc. and by ligation of other cell surface receptors. The ~1 integrin consists of a common ~1 subunit (CD 29) noncovalently linked to a a subunit and are found on nearly all cell types. However, one ~l-integrin, VLA-4, is found primarily on leukocytes (Werr et al., 1998). It is constitutively expressed on all circulating leukocytes except PMN and like ~2 integrin requires activation in order to bind. The ~7 integrin is expressed on some Band T lymphocytes and functions as the homing receptor for intestinal lymphoid tissue (Carlos & Harlan, 1994). Immunoglobulin Superfamily Members of the immunoglobulin superfamily constitute the third class of adhesion molecules. The superfamily consists of cell surface proteins that
Inflammation and Mediators
169
are involved in antigen recognition and complement binding, as well as cell adhesion. Endothelial members of this superfamily involved in leukocyte endothelial cell adhesion.
Adhesion Cascade and Inflammation The sequence of events involved in leukocyte adherence to vascular endothelium and transmigration into tissue is complex and has been termed the adhesion cascade (Springer, 1994). The first step involves a random contact of the leukocytes with the endothelial cell surface. After initial contact, relatively weak binding of L, P, and/or E selectin with their carbohydrate ligands occurs. Under conditions of flow, these multiple weak interactions are sufficient to slow the leukocyte down, and the leukocyte is seen to roll along the surface of the endothelial cells. The next involves further activation of the endothelial cells and leukocytes by cytokines, chemoattractants, and chemokines that are produced locally (Berlin et al., 1995). This leads to the upregulation and/or activation of the leukocyte integrins and the endothelial Ig-like molecules. The interaction of these two classes of adhesion molecules mediates the firm adherence ofleukocytes to the endothelial cell surface (Huber et al., 1991). The firmly adherent leukocytes migrate across the endothelial cell surface via integrin-Ig like interactions, diapedesis between endothelial cells and then migrate to the sub endothelial matrix via integrin binding to matrix components. (Feng et al., 1998). While the adherence cascade functions normally to recruit leukocytes to extravascular sites for host defense and repair, under some circumstances leukocytes are firmly adhere to the endothelium, a protected microenvironment is found. In this microenvironment, inflammatory mediators produced by the leukocytes may potentially reach high concentrations and overcome local and systemic antiinflammatory protective mechanisms, thereby allowing endothelial cell injury to occur resulting in an increase in microvascular permeability and hemorrhage. This series of events may initiate and sustain a cycle of inflammation, leading to further leukocyte recruitment and endothelial cell injury. The continued recruitment of leukocytes can lead to occlusion of the microvasculature by leukocyte aggregates, causing local ischemia. The leukocytes may also diapede between endothelial cells, gaining access to the extravascular space, where they can mediate further tissue damage, producing organ dysfunctions. This cascade of events leading to vascular and tissue injury dependent on leukocyte-endothelial adhesive interactions, and thus, interrupting the interactions at the level of rolling, firm adherence, or diapedesis should decrease leukocyte mediated injury, in other words inflammation. In addition to inflammation, integrins and selectins are also involved in other abnormal conditions, such as septic shock, thrombosis, reperfusion damage or metastasis (Frenette & Wagner, 1996; Mojcik & Shevach, 1997; Gonzalez-Amaro et al., 1998).
170
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Phagocytosis and Inflammation When inflammation remains unresolved, stimulated phagocytes can inflict damage on tissues and produce superoxide anion (0-2) which in turn forms extremely toxic metabolites such as the hydroxyl radical (O-H), peroxy radical (O-OH) hypochlorous acid, hydrogen peroxide, singlet oxygen and hydroperoxide (Rowe et al., 1983). All these oxygen species derived from superoxide are damaging either by a powerful, direct oxidizing action or indirectly as with the hydroxyl and perhydroxyl radical which initiates lipid per oxidation and consequently results in membrane destruction (Mead, 1976). Tissue damage provokes inflammatory responses by the production of mediators and chemotactic factors. Nature has programmed phagocytes such as the neutrophil to use these toxic oxygen metabolites to kill invading organisms. Phagocytic cell attaches itselfto the foreign particle at certain specific sites. These sites also have an adjacent enzyme system known as NADPH-oxidase (Roos et al., 1976) which converts oxygen to superoxide anion which in turn is converted to other toxic oxygen species. Moreover, phagocytes are not particularly sensitive in identifying microorganisms. In general, they will attach to any surface which they are unable to recognize as 'self. In rheumatoid arthritis, the invading neutrophils attach themselves to collagen fibrils in cartilage and other joint tissues (Bromley et al., 1984; Crisp et al., 1984). These fibrils have been made 'non self to the neutrophils by considerable biochemical damage inflicted during the inflammatory process. The phagocyte is physically incapable of engulfing these large structures but attempts to do so. A vesicle is opened up on the surface of the phagocyte but the massive size of the collagen prevents the vesicle from closing and it remains open to the extracellular compartment. However, the cell is stimulated and lysosomes fuse with the vesicle and superoxide anion and other oxygen products are produced. These products bathe the collagen fibrils and other structures with active oxygen species and hydrolytic enzymes. More tissue is rendered 'non-self by the damage inflicted. Consequently more phagocytes are attracted by chemotaxis in an attempt to remove it and a cascade of destruction is initiated. The nonself material is also the source of auto-antigens, characteristic of chronic inflammation (Scher et al., 1980). Therefore, the blood of patients with rheumatoid arthritis contains large number of antibodies which have been formed to act against auto-antigens produced by enzymic or oxygen damage. A characteristic antibody is rheumatoid factor is formed which is an antibody formed against altered immunoglobulins (IgG). Another common antibody formed is antinuclear antibody (ANA), formed against antigens produced from cells disrupted in the disease process. The phenomenon of frustrated phagocytosis is a key pathological event in the transformation of acute inflammation to chronic inflammation.
Inflammation and Mediators
171
CONCLUSIONS The process of inflammation is very complex; different scientists have given different views regarding inflammation and its propagation. To understand the complexity of this process we tried to explain some of the mediator mechanisms of this protective defence of the body. We feel that this review shell give some understanding to the readers about the basis of inflammation and its mediators responsible for its initiation and activation yet this is not the ultimate according to our views but some of the achievements of the science towards inflammation. Although there are still so many things left unwound to know completely about inflammation yet a sincere attempt has been made to explain the phenomenon of inflammation and its mediators.
REFERENCES Abramson, S.B. and Wissmann, G. (1989). The mechanism of action of non-steroidal antiinflammatory drugs. Arth. Rheum., 32: 1-9. Allcan, 1. and Kubes, P.A. (1996), Critical role for nitric oxide in intestinal barrier function and dysfunction. Am. J. Physiol., 270: G225-237. Alnemri, E.S. (1997). Mammalian cell death proteases: A family of highly conserved asparate specific cysteine proteases. J. Cell Biochem., 64: 33-42. Alvarellos, A., Lipsky, P.E. and Jasin, H.E. (1988). Prostaglandin E2 modulation of rheumatoid factor synthesis. Arth. Rheum., 7: 1473-1480. Anderson, A.a., Anderson, N.D. and White, J.D. (1982). Lymphocyte locomotion, lymphocyte tissue and lymphocyte circulation in the rat. In: Animal models of immunological processes. J.B. Hay, Eds., pp 25-95, Academic press, New York. Aplin, A.E., Howe, A., Alahari, S.K. and Juliano, RL. (1998). Signal transduction and signal modulation by cell adhesion receptors: the role of integrins, cadherins, immunoglobulin-cell adhesion molecules and selectins. Pharmacol. Rev., 50: 199-263. Arrang, J.M., Garbarg, M. and Schwartz, J.C. (1983). Autoinhibition of brain histamine release mediated by a novel class (H3) receptor. Nature, (Lond.) 302: 832-837. Ash, A.S.F. and Schild, H.O. (1966), Receptors mediating some action of histamine. Br. J. Pharmacol., 27: 427-439. Bautista, A.P. and Spitzer, J.J. (1994). Inhibition of nitric oxide formation in vivo enhances superoxide release by the perfused liver. Am. J. Physiol., 266: G783-G788. Beauerle, P. and Baltimore (1996). NF-KB: Ten years after. Cell, 87: 13-20. Berlin, C., Bargatze, RF., Campbell, J.J., von Andrian, V.M., Szabo, M.C., Hasslen, S.R, Nelson, RD., Berg, E.L., Erlandsen, S.L. and Butcher, E.C. (1995). a 4 integrins mediate lymphocyte attachment and rolling under physiologic flow. Cell, 80: 413-422. Bjorkman, D.J. (1998b). The effect of aspirin and non-steroidal antiinflammatory drugs on prostaglandins. Am J. Med., 105(1B): 8S-12S. Blachier, F., Mrabet-Touil, H., Darcy-Villon, B., Posho, 1. and Duee, P.H. (1991). Stimulation by D-glucose of the direct conversion of arginine to citrullin in enterocytes isolated from pig jejunum. Biochem. Biophy. Res. Commun., 177: 1171-1177. Black, J.W., Duncan, W.A.M., Durant, C.J., Ganellin, C.R and Parsons, E.M. (1972). Definition and antagonism of histamine H2 receptors. Nature (Lond), 236: 385-390. Bogle, RG., Baydoun, A.R, Pearson, J.D., Moncada, S. and Mann, G.E. (1992). Arginine transport is increased in macrophage generating nitric oxide. Biochem. J., 284: 5-18. Bolten, W.W. (1998). Scientific rationale for specific inhibition of COX-2. J. Rheumatol., 25(suppl): 2-7.
172
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Brain, S.D., Camp, RD., Dowd, P.M. and Greaves, M.W. (1985). Quoted In: New perspective of non-steroidal antiinflammatory drugs, an experimental study.B-Pharma thesis Leena Raj, Panjab University Chandigarh (1990). Bray, M.A. (1987). Prostaglandins and leukotrienes: fine tuning the immune response. lSI Atlas of Science: Pharmacology, 1: 101-106. Bray, M.A., Cunningham, F.M., Ford-Hutchinson, A.W. and Smith, M.J.H. (1981). Leukotrien B 4 : a mediator of vascular permeability. Br. J. Pharmacol., 72(3): 483-486. Bredt, D.S., Glatt., C.E., Huang, P.M., Dawson., T.M. and Snyder, S.H. (1991). Nitric oxide synthase protein and mRNA are discreetly localised in neuronal populations of mammalian CNS together with NADPH diaphorase. Neuton, 7: 615-624. Bredt, D.S. and Snyder, S.H. (1990). Isolation of nitric oxide synthase, a calmodulin requiring enzyme. Proc. Natl. Acad. Sci., 87: 323-328. Bruch Gerharz, D., Fehsel, K., Suschek, C., Michal, G. and Ruzicka, T.A. (1996). Proinflammatory activity of interleukin-8 in human skin: expression of the inducible nitric oxide synthase in psoriatic lesions and cultured keratinocytes. J. Expt. Med., 184: 2007-2012. Brombley, M., Fischer, W.D. and Wolley, D.E. (1984). Mast cells at site of cartilage erosion in the rheumatoid joint. Ann. Rheumat. D~s., 43: 76-79. Brooks, P.M. and Day, RO. (1991). Non-steroidal antiinflammatory drugs differences and similarities. New Engl. J. Med., 324: 1716-1725. Buss, W.W. (1979). Histamine: Mediator and modulator in inflammation. In: Houck, J.C. (ed.): Chemical messengers of the inflammatory process. Amsterdam, Elsevirel North Holland. P-1. Butcher, E.C. (1991). Leukocyte-endothelial cell recognition: three (or more) steps to specificity and diversity. Cell, 67: 1033-1036. Camp, RD.R, Chu, A, Cunningham, F.M., Fincham, N.J., Greaves, M.W. and Morris, J. (1986). Identification of an interleukine-l like peptide in psoriasis. Br. J. Pharmacol., 88: 244B. Cassatella, M.A (1995). The production of cytokines by polymorphonuclear neutrophils. Immunol. Today, 13: 69-72. Cassatella, M.A. (1999). Neutrophil-derived proteins: Selling cytokines by the pound. Adv. Immunol., 73: 369-509. Cassone, A, Chiani, P., Quinti, I. and Torosantucc, A (1997). Possible participation of polymorphonuclear cells stimulated by microbial immunomodulators in the dysregulated cytokine pattern of AIDS patients. J. Leukoc. Biol., 62: 60-66. Campion, G.V., Lebsack, M.E., Lookabaugh, J., Gordon, G. and Catalano, M. (1996). Dose range and dose frequency study of recombinant human, interleukin-l receptor antagonist in patients with rheumatoid arthritis. The IL-l IRa arthritis study group. Arthritis Rheum., 39: 1092-1102. Carlos, T.M. and Harlan, J.M. (1994). Leukocyte-endothelial adhesion molecules. Blood, 84: 2068-2101. Charles, I.G., Palmer, RM.J., Hickery, M.S., Bayliss, M.T., Chubb, AR, Hall, V.S., Moss, D.W. and Moncada, S. (1993). Cloning characterization and expression of eDNA encoding an inducible nitric oxide synthase from the human chondrocyte. Proc. Natl. Acad. Sci., 90: 11419-11423. Cheronis, J.C., Whalley, E.T., Nguyen, K.T., Eubanks, S.R and Allan, L.G. (1992). A new class of bradykinin antagonist: Synthesis and in vitro activity of Bissuccinimdoalkane peptide dimers. J. Med. Chem., 35: 1563-1572. Colville-Nash, P.R. and Scott, D.L. (1992). Angiogenesis and rheumatoid arthritis; pathogenic and therapeutic implications. Ann. Rheum. Dis., 51: 919-925. Corraliza, I.M., Campo, M.L., Fuentes, J.M., Campos-portuguez, S. and Soler, G. (1993). Parallel induction of nitric oxide and glucose-6 phosphate dehydrogenase in activated bone marrow derived macrophages. Biochem. Biophy. Res. Commun., 196: 343-347. Crisp, AJ., Chapman, C.M. and Krane, S.M. (1984). Articular mastocytosis in rheumatoid arthritis. Arth. Rheum., 27: 845-851.
Inflammation and Mediators
173
Cush, J.J. and Kavanaugh, F.A (1995). Biological interventions in rheumatoid arthritis. Rheum. Dis. Clin. N. Am., 99: 82-88. Dawson, J., Edwords, J.C.W., Sedgwick, AD. and Lees, P. (1991). IL-1a inhibits lymphocyte migration into a site of chronic inflammation. Immunol Letters, 30: 319-324. DeClerk, F.F. and Harman, AR (1983). 5-hydroxytryptamine and platelet aggregation. Fed. Proc., 42: 228-232. DeWitt, D.L. (1995). The differential susceptibility of prostaglandin endoperoxide H synthase1 and -2 to non-steroidal antiinflammatory drugs: aspirin derivatives as selective inhibitors. Med. Chem. Res., 5: 325-343. Dinarello, C. (1994). The interleukin-1 family: 10 years of discovery. FASEB. J., 8: 13141335. Dinarello, C. and Wolff, S. (1993). The role of interleukin-1 in disease. N. Engl. J. Med., 328: 1513-1518. Djeu, J.y', Serb Ousek, D. and Blancherard, D.K (1990). Release of tumor necrosis factor by human polymorphonuclear leukocytes. Blood, 76: 1405-1409. Drevlow, B.E., Lovis, R, Haag, M.A, Sinacore, J.M., Jacobs, C., Blosche, C., Landay, A, Moreland, L.W. and Pope, RM. (1996). Recombinant human, interleukin-1 receptor type1 in the treatment of patients with active rheumatoid arthritis. Arthritis Rheum., 39: 257-265. Dray, A and Perkins, M.N. (1993). Bradykinin and inflammatory pain. Trends Neuroscl., 16: 99-104. Dray, A. and Urban, L. (1996). New pharmacological studies for pain relief. Ann. Rev. Pharmacol. Toxicol., 36: 253-280 Dunon, D., Mackay, C.R and Imof, B.A. (1993). Adhesion in leukocyte homing and differentiation. pp 260, Springer verlag, New York. Elford, P.R, Heng, R, Revesz, L. and MacKenzi (1995), Reduction of inflammation and pyrexia in the rat by oral administration ofSDZ-224-015, an inhibitor ofinterleukin-1b converting enzyme. Br. J. Pharmacol., 115: 601-606. Farmer, S.G. (1995), Bradykinin, in airways smooth muscle: Peptide receptors, ion channels and signal transduction (Raeburn, D. & Gimbyez, M.A. eds.) pp. 51-65. Birkbauser, Basel. Fava, RA, Olsen, H.J., Postlethwaite, A.E., Broadley, KN., Davidson, J.M., Nanney, L.B., Lucas, C. and Townes, AS. (1991). Transforming growth factor b1 induced neutrophil recruitment to synovial tissue: Implications for transforming growth factor b driven synovial inflammation and hyperplasia. J. Exp. Med., 173: 1121-1132. Feldman, P.F., Griffith, O.F. and Stuehr, D.J. (1993). The surprising life of nitric oxide. Chem. Eng. News, 71(51): 26-39. Ferreira, S.H. and Vane, J.R (1979). Mode of action of antiinflammatory agents which are prostaglandin synthetase inhibitors. In: Vane, J.R and Ferreira, S.H. (eds.), Antiinflammatory Drugs: Handbook of experimental pharmacology, 50111: pp. 348-393 Springer Verlag Berlin New York. Arend, W.P. and Dayer, J.M. (1990). Cytokine and cytokine inhibitors or antagonists in rheumatoid arthritia. Arth. Rheum., 33: 305-315. Farrell, A.J., Blake, D.R, Palmer, RM. and Moncada, S. (1992). Increased concentration of nitrite in synovial fluid and serum samples suggest increased nitric oxide synthesis in rheumatic diseases. Ann. Rheum. Dis., 51: 1219-1222. Fawcell, D.W. (1994). "A textbook of histology", 12th edn., pp 964 Champman and Hall, New York. Feng, D., Nagy, J.A., Pyne, K, Dvorak, H.F. and Dvorak, AM. (1998). Neutrophils emigrate from venules by a transendothelial cell pathway in response to FMPL. J. Exp. Med., 187: 903-915. Firestein, G. and Zvaifler, N. (1997). Anticytokine therapy in rheumatoid arthritis. N. Engl. J. Med., 337: 195-197. Fraga, C.G., Shignaga, M.K., Park, J.W., Degan, P. and Ames, B.N. (1990). Oxidative damage to DNA during aging: 8-hydroxy-2-deoxyguanosin in rat organ DNA and urine. Proc. Natl. Acad. Sci. USA., 87: 4533-4537.
174
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Frenette, P.S. and Wagner, D.D. (1996). Adhesion molecules- partlI. Blood vessels and blood cells. N. Engl. J. Med., 335: 43-45. Frolich, J.C. (1997). A classification of NSAIDs according to the relative inhibition of cyclooxygenase isoenzymes. Trends Pharmacol. Sci., 18: 30-34. Galen, (1978). Quoted In: Vane, J.R. and Ferreira, S.H. (eds.), Antiinflammatory Drugs: Handbook of experimental pharmacology, 50111: pp. 1-4 Springer Verlag Berlin New York Gaston, B., Reilly, J. and Drazen, J.M. (1993). Endogenous nitric oxide and bronchodilators S-nitrosothiols in human airways. Pros. Nat!. Acad. Sci. USA., 90: 10957-10961. Gonzalez-Amaro, R., Diaz-Gonzalez, F. and Sanchez-Madrid, M. (1998). Adhesion molecules in inflammatory diseases. Drugs, 56: 977-988. Goodwin, J.S. (1991). Are prostaglandins proinflammatory, antiinflammatory, both or neither? J. Rheumatol., 18(suppl): 26-29. Gu, Y., Kuida, K, Tsutsui, H., Ku, G., Hsiao, K, Fleming, M.A., Hayoshi, N., Higashino, K, Okamura, H., Nakanishi, K, Kurimoto, M., Tanimoto, T., Flavell, R.A., Sato, V., Harding, M.W., Livingstone, D.J. and Su, M.S.S. (1997), Activation of interferon-g inducing factor mediated by interleukin-lb converting enzyme. Science, 275: 206-209. Hall, J.M. (1997). Bradykinin receptors: Gen. Pharmacol., 28: 1-6. Halliwell, B. and Gutteridge, J.M.C. (1985), The importance of free radicals and catalytic metal ions in human diseases. Mol. Asp. Med., 8: 89-193. Harris, G., Bashir, S. and Winyard, P.G. (1994). 7,8-dihydro-8-oxo-2- deoxyguanosin present in DNA is not simply an artefact of isolation. Carcinogenesis, 15: 411-415. Hartwell, D.W., Mayadas, T.N., Berger, G., Frenette, P.S., Rayburn, H., Hynes, R.O. and Wagner, D.D. (1998), Role of P-selectin cytoplasmic domain in granular targeting in vivo and in early inflammatory response. J. Cell Bio!., 143: 129-1141. Heapy, C.G., Shaw, J.S. and Farmer, S.C. (1993). Differential sensitivity of antinociceptive assays to bradykinin antagonist Hoe 140. Br. J. Pharmacol., 108: 209-213. Hibbs, J.B. Jr., Taintor, R.R. and Vavrin, Z. (1987). Maccrophage cytotoxicity: role of Larginine deiminase and immuno nitrogen oxidation to nitrite. Science, 235: 473-476. Hla, T. and Neilson, K (1992), Human cyclooxygenase-2 cDNA. Proc. Natl. Acad. Sci. USA., 89: 7384-7388. Hock, F.J., Wirth, K, Albus, u., Linz, W. and Gerhards, H.S. (1991). HOE -140, a new potent long acting bradykinin antagonist in vitro studies. Br. J. Pharmacol., 102: 769774. Houglum, K, Bedossa, P. and Chojkier, M. (1994). TGF- beta and collagen-alpha 1 (I) gene expression are increased in hepatic acinar zone 1 of rats with iran overload. Am. J. Physiol., 267: G908-G913. Huber, A.R., Kunkel, S.L., Todd, R.F. and Wiss, S.J. (1991). Regulation of transendothelial neutrophil migration by endogenous interleukin-8. Science, 254: 99-102 Hynes, R. (1992). Integrins, versatility, modulation and signaling in cell adhesion. Cell, 69: 11-25. Ialenti, A., Ianaro, A., Moncada, S. and DiRosa, M. (1992). Modulation of acute inflammation by endogeneous nitric oxide. Eur. J. Pharmacol., 211: 177-182. Inove, Y., Bode, B.P., Beck., D.L., Li, A.P., Bland, KI. and Souba, W.W. (1993). Arrginine transport in human liver. Characterization and effect of nitric oxide synthase inhibitors. Ann. Surg., 218: 350-363. Jacobson, M.D. (1996). Reactive oxygen species and programmed cell death. TIBS., 21: 83-86. Kajeker, R., Moore, P.K and Brain, S.D. (1995). Essentiol role of nitric oxide in neurogenic inflammation in rat cutaneous microcirculation. Evidence for an endothelium independent mechanism. Circ. Res., 76: 441-447. Keaney, J.F. Jr., Simon, D.I., Stamler, Jaraki, 0., Scharfstein, J., Vita, J.A. and Loscalzo, J. (1993). NO forms an adduct with serum albumin that has endothelium derived factor like properties. J. Clin. Invest., 91: 1582-1589. Kharitonov, S.A., Sapienza, M.M., Chung, KF. and Barnes, P.J. (1999). Prostaglandins mediate bradykinin-induced reduction of exhaled nitric oxide in asthma. Eur. Respir. J., 14: 1023-1027.
Inflammation and Mediators
175
Kindgen Milles, D. and Arndt, J.O. (1996). Nitric oxide as a chemical link in the generation of pain from veins in humans. Pain, 64: 139-142. Knowles, R.G., Placios, M., Palmer, R.M. and Moncada, S. (1989). Formation of nitric oxide from L-argenine in the central nervous system: a transduction mechanism for stimulation of the soluble guanylate cyclase. Proc. Natl. Acad. Sci., 86: 5159-5162. Kristal, B.S., Kim, J.D. and Yu, B.P. (1994). Tissue-specific susceptibility to peroxy redicalmediated inhibition of mitochondrial transcription. Redox Res., 1: 51-55. Ku, G., Faust, T., Lauffer, L.L., Livingstone, D.J. and Harding, M.H. (1996). InterleukinIb converting enzyme inhibition blocks progression of collagen type-II induced arthritis in mice. Cytokine, 8: 377-389. Kujubu, D.A., Fletcher, B.S., Varnum, B.C., Lim, R.W. and Herschman, H.R. (1991). TISI0, a pharbol ester tumor promotor-inducible mRNA from Swiss 3T3 cells, enclosed a novel prostaglandin synthase/ cyclooxygenase homolog. J. Biol. Chem., 266: 1286612872. Kuida, K., Lippke, J., Ku. G., Harding, M.H., Livingstone, D., Su, M. and Flavell, R. (1995). Altered cytokine export and apoptosis in mice defecient in interleukin-lb converting enzyme. Science, 267: 2000-2002. Kus, M.L., Fairburn, K., Blake, D.R. and Winyard, P.G. (1995). A vascular basis for free radical involvement in inflammatory joint disease. In: Immunopharmacology of freeredical species. Blake, D. and Winyard, P.G (eds.), pp. 97-112 Academic Press London. Leone, A.M., Palmer, R.M., Knowles, R.G., Francis, P., Ashton, D.S. and Moncada, S. (1991). Constitutive and inducible nitric oxide synthases incorporate molecular oxygen into both nitric oxide and citrulline. J. Biol. Chem., 266: 23790-23795. Lovenberg, T.W., Roland, B.L., Wilson, S.J., Jiang, X., Pyati, J., Huver, A., Jackson, M.R. and Erlander, M.G. (1999). Cloning and functional expression of the human histamine H3 receptor. Mol. Pharmacol., 55: 1101-1107. Lord, P.C., Wilmoth, L.M., Mizel, S.B. and McCall, C.E. (1991). Expression of interleukinla and ~ genes by human blood polymorphonuclear leukocytes. J Clin. Inves., 87: 1312-1321. Lind, D.S., Copeland, E.M. and Souba, W.W. (1993). Endotoxin stinulates arginine transport in pulmonary artery endothelial cells. Surgery, 11: 4199-4205. Liu, C., Ma, X.J., Jiang, X., Wilson, S.J., Hofstra, C.L., Blevitt, J., Pyati, J., Li, X., Chi, W., Curruthera, N. and Lovenberg, T.W. (2001). Cloning and pharmacological characterization of a fourth histamine receptor (H4) expressed in bone marrow. Mol. Pharmacol., 59: 420-426. Lorsbach, R.B., Murphy, W.J., Lowenstein, C.J., Synder, S.H. and Russel, S.W. (1993). Expression of nitric oxide synthase gene in mouse macrophages activated for tumor cell killing. Molecular basis for the synergy between interferon-y and lipopolysaccharide. J. Biol. Chem., 268: 1908-1913. Malmberg, A.B. and Yaksh, T.L. (1992). Hyperalgesia mediated by spinal glutamate or substance P receptor blocked by spinal cyclooxygenase inhibitor. Science, 257: 12761279. MarIetta, M.A. (1991). Nitric oxide synthase structure and mechanism. J. Biol. Chem., 268: 12231-12234. Masferrer J.L., Isakson, P.C. and Seiber, K. (1996). Cyclooxygenase-2 inhibitor, a new class of antiinflammatory agents that spare the gastrointestinal tract. Gastroentrology Clin. North Am., 25: 363-372. Mauke, J.G., Borkowski, J.A., Bierilo, K., MackNeil, T. and Derrick, A.W. (1994). Expression cloning of a human Bl bradykinin receptor. J. Biol. Chem., 269: 2158321586. McEver, R.P. (1994). Selectins. Curro Opin. Immunol., 6: 75-84. Mead, J.F. (1976). Free redical mechanism of lipid damage and consequences for cellular membranes. Free radicals in biology (ed.) Pryor, W.A. New York Academic Press, 1: 51-68. Merry, P., Winyard, P.G., Morris, C.J., Grootvetd, M. and Blake, D.R. (1989). Oxygen free radicals, inflammation and synovitis: the current status. Ann. Rheum. Dis., 48: 864870. Miossec, P. and van der Berg, W. (1997). Thl/ Th2 cytokine balance in arthritis. Arthritis rheum., 40: 2105-2115.
176
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Miura, M., Zhu, H., Rotello, R, Hartwieg, E.A. and Yuan, J. (1993). Induction of apoptosis in fibroblasts by IL-113 converting enzyme, a mammalian homologue of the C. elegans cell death gene Ced-3. Cell, 75: 653-660. Mojcik, C.F. and Shevach, W.W. (1997). Adhesion molecules. A rheumatologic perspective. Arthritis Rheum., 40: 991-1004. Moncada, S., Palmer, R.M. and Higgs, E.A. (1990). Relationship between prostacyclin and nitric oxide in the thrombotic process. Throm. Res. Suppl., 11: 3-13. Moncada, S., Palmer, R.M.J. and Higgs, E.A. (1991). Nitric oxide physiology, pathophysiology and pharmacology. Pharmacol. Rev., 43: 109-142. Murphy, S., Simmons, M.L., Agullo, L., Garcia, A., Feinstein, D.L., Galea, E., Reis, D.J., Minc-Golomb, D. and Schwartz, J.P. (1993). Synthesis of nitric oxide in CNS glial cells. Trends Neur. Sci., 16: 323- 328. Nakamura, T., Itadani, H., Hidaka, Y., Ohta, M. and Tanaka, K. (2000). Molecular cloning and characterization of a new human histamine receptor, HH4R Biochem. Biophys. Res. Commun., 279: 615-620. Nissler, A.K. and Billar, T.R (1993). Inflammation, immunoregulation and inducible nitric oxide synthase. J. Leukocyte Biol. 54: 171-178. Palmer, R.M.J., Aston, D.S. and Moncada, S. (1988). Vascular endothelial cells synthesize nitric oxide from L-arginin. Nature (London), 333: 664-666. Perkins, M.N., Campbell, E. and Dray, A. (1993). Antinociceptive activity of the bradykinin Bland B2 receptor antagonists, des-Arg9, (Le48)-Bk and HOE 140 in two models of persistent hyperalgesia in the rat. Pain, 53: 191-197. Peskar, B.M. and Maricic, N. (1998). Role of prostaglandins in gastroprotection. Dig. Dis. Sci., 43(suppl): 23S-29S. Phipps, RP., Stein, S,H. and Roper, RL. (1991). A new view of prostaglandin E regulation of the immune response. Immunol. Today, 12: 349-352. Piper, P.J. and Vane, J.R (1971). The release of prostaglandins from lungs and other tissues. Ann. N. Y. Acad. Sci., 180: 363-383. Rapport, M.M., Green, A.A. and Page, I.H. (1948). Serum vasoconstrictor (serotonin) isolation and characterization. J. Biol. Chem., 176: 1243-1251. Richardson, C.E. and Emery, P. (1995). New cyclooxygenase and cytokine inhibitors. Baillieres Clin. Rhumatol., 9: 731-758. Richter, C., Park, J.W. and Ames, B.N. (1988). Normal oxidative damage to mitochondrial and nuclear DNA is extensive. Proc. Natl. Acad. Sci. USA., 85: 6465-6467. Robinson, D.W. (1991). Eicosanoids in human inflammation. In: hedqvist, P., Kalden, J.R, Muller-Peddinghaus, R and Robinson, D.R, (eds.) Trends in RA-research, Basel: EULAR, 195-198. Roos, D.J., Hamoun-Muller, W.T. and Weening, RJ. (1976). Effect of cytochalosin-B on the oxidative metabolism of human peripheral blood granulocytes. Biochem. Biophys. Res. Commun., 68: 43-50. Rosen, S.D. and Bertozzi, C.R (1994). The selectins and their ligands. Curro Opin. Cell Biol., 6: 663-673. Rothwell, N.J. (1992). Eicosanoids, thermogenesis and thermoregulation. Prostaglandins, Leukotrienes and Essential Fatty Acids, 46: 1-7. Rowe, G.T., Manson, N.H., Caplan, M. and Hess, M.L. (1983). Hydrogen peroxide and hydrogen radical mediation of activated leukocyte depression of cardiac sarcoplasmic reticulum. Cir. Res., 53: 584-591. Rubbo, H., Raid, R, Trujillo, M., Telleri, R, Kalyanaraman, B., Barnas, S., Kirk, M. and Ferrman, B.A. (1995). Nitric oxide regulation of superoxide and peroxynitrite-dependent lipid peroxidation: formation of noval nitrogen-containing oxidized lipid derivatives. J. Biol. Chem., 269: 26066-26075. Safayhi, S.D., Mack, T., Sabiraj, J., Michael, LA., Subramanian, L.R and Ammon, H.P.T. (1992). Boswellic acids: Noval, specific nonredox inhibitor of 5-lipoxygenase. J. Pharmac. Exp. Ther., 261(3): 1143-1146. Salvesen, G.S. and Dixit, V.M. (1997). Caspases: Intracellular signaling by proteolysis. Cell, 91: 443-446.
Inflammation and Mediators
177
Saudou, F. and Hen, R (1994). 5-hydroxytryptamine receptor subtypes: molecular and functional diversity. Adv. Pharmacol., 30: 327-380. Sawutz, D.G., Salvino, J.M., Dolle, RE., Casianio, F. and Ward, S.J. (1994). The nonpeptide WIN 64388 is a bradykinin B2 receptor antagonist. Proc. Natl. Acad. Sci. USA., 91: 4693-4697. Scher, M.G., Beller, D.L. and Unanue, E.R (1980). Demonstration of a soluble mediator that induces exudates rich in macrophages. J. Expt. Med., 152: 1684. Simon, L.S., Lanza, F.L. and Lipsky, P.E. (1989). Preliminary study of safety and efficacy of SC-58635, a novel cyclooxygenase-2 inhibitor. Arth. Rheum., 41: 1591-1602. Singh, G.B. (1983), Pharmacological investigations of some newly developed alkyl and aryl stryl ketones. M. Phil Thesis submitted, University Of Bradford. pp. 4-36. Sleight, A.J., Pierce, P.A., Schmidt, A.W., Hekmatpanah, C.R and Peroutka, S.J. (1991). The clinical utility of serotonin receptor activity agent in neuropsychiatric disease. In: Serotonin receptor subtype: basic and clinical aspects. S. Peroulka (ed.). pp. 147-210. John Wiley and Sons, New York. Springer, T.A. (1994). Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigma. Cell, 76: 301-314. Stadler, J., Stefanovic-Racic, M., Billiar, T.R, Curran, RD., Mclntyre, L.A., Georgescu, H.I., Simmons, L.R and Evans, C.H. (1991). Articular chondrocytes synthesize nitric oxide in response to cytokines and lipopolysaccharide. J. Immunol., 147: 3915-3917. Sterauka, RR., Manning, D., DeHass, C.J., Ferkany, J.W. and Borosky, A.S. (1988). Bradykinin as a pain mediator: receptors are located to sensory neurons and antagonists have analgesic action. Proc. Natl. Acad. Sci. USA., 85: 3245-3249. Stefanovic-Racic, M., Stadler, J. and Evans, C.H. (1993). Nitric oxide and arthritis. Arth. Rheum., 36: 1036-1041. Stuehr, D.J. and Griffith, O.W. (1992). Mammalian nitric oxidesynthase. Adv. Enzy. Relat. Areas Mol. Biol., 65: 287-346. Twarog, B.M. and Page, J.H. (1953). Serotonin content of some mammalian tissue and urine and a method for detection. J. Physiol., 175: 157-161. Vane, J.R (1971). Inhibition of prostaglandin synthesis as a mechanism of action of aspirin-like drugs. Nature, 231: 232-235. Vestweber, D. and Blank, J.E. (1999). Mechanism that regulate the function of the selections and their ligands. Physwl. Rev., 79: 181-213. Vialli, M. and Erspamer, V. (1933). Cellule enterocromaffinie cellule basigranulose acidofile nei vertebati. Z. Zellforsch. Mikrosk Anat., 19: 743. Vodovotz, Y., Bogdam, C., Paik, J., Xie, Q.W. and Nathan, C. (1993).Mechanism of suppression of macrophage nitric oxide release by transforming growth factor-~. J. Expt. Med., 178: 605-613. Wang, S., Miura, M., Jung, Y.K., Zhu, H., Li, E. and Yuan, J. (1998). Murine caspase-11, an ICE-interacting protease, is essential for the activity of the ICE/ Ced-3 family and an upstream regulator of ICE. Cell, 92: 501-509. Watson, S.R. and Bradlley, L.M. (1998), The recirculation of naIve and memory lymphocytes. Cell adhesion Commun., 6: 105-110. Weigert, C. (1889). Cited In: Metchnikoff, E. Lecons, sur la Pathologie camparee de l' inflammation, Masson, Paris (1892). Werr, J., Xie, X., Hedqvist, P., Ruoslahti, E. and Lindbom, L. (1998). ~1 integrins are critically involved in neutrophil locomotion in extravascular tissue in vivo. J. Exp. Med., 187: 2091-2096. Wilkinson, L.O. and Dourish, C.T. (1991). Serotonin and animal behavior. In: Serotonin receptor subtype: basic and clinical aspects. S. Peroulka (ed.). pp. 147-210 John Wiley and Sons, New York. Williams, K.I. and Higgs, G.A. (1988). Ecosanoids and inflammation. J. Pathology, 156: 101-110. Willis, A. L. (1969). Release of histamine, kinin and prostaglandin during carrageenaninduced inflammation in rats. In: Mantegazza, P. and Horton, E.D. (eds.): Prostaglandins, pep tides and amines, pp 33-38 London - New York, Academic press.
178
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Wissman, I.L. and Cooper, M.D. (1993). How the immune system develops. Sci. Am., 269(3): 33-39. Xie, Q.W., Cho, H.J., Calayca, Y.J., Mumford, R.A., Swiderek, KM., Lee, T.D., Ding, A., Troso, T. and Nathah, C. (1992). Cloning and characterization of inducible nitric oxide synthase from mouse macrophages. Science, 256: 225-228.
7 Recommendations for Reporting Randomized Controlled Trials of Herbal Interventions: CONSORT for Herbal Medicine Trials JOEL J. GAGNIER N.D.l*, HEATHER BOON 2, PAULA ROCHON 3 , DAVID MOHER4, JOANNE BARNEs5 AND CLAIRE BOMBARDIER6 FOR THE CONSORT GROUP
ABSTRACT Controlled trials that utilize randomized allocation are the best tool to control for bias and confounding in trials testing clinical interventions. Investigators must be sure to include in reports of these trials information that is required by the reader to judge the validity and implications of the findings. In part, complete reporting of trials will allow clinician's to modify their clinical practice to reflect current evidence towards the improvement of clinical outcomes. The CONSORT statement was developed to assist investigators, authors, reviewers and editors on the necessary information to be included in reports ofcontrolled clinical trials. The CONSORT statement is applicable to any intervention, including herbal medicinal products. Controlled trials of herbal interventions do not adequately report the information suggested in CONSORT. Recently, reporting recommendations were developed in which several CONSORT items were elaborated to become relevant and complete for randomized controlled trials of herbal medicines. We expect 1. 5955 Ontario St, Unit 307, Windsor, Ontario, Canada, N8S1W6. 2. Leslie Dan Faculty of Pharmacy, 19 Russell Street, University of Toronto, Toronto, Ontario, Canada, M5S 2S2. 3. Baycrest Centre, 3560 Bathurst Street, Toronto, Ontario, Canada, M6A 2El. 4. Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Rm. 210, Ottawa, Ontario, Canada, K1H 8L1. 5. School of Pharmacy, University of Aukland. Position:. Associate Professor in Herbal Medicines. 6. Institute for Work and Health, 481 University Avenue, Suite 800, Toronto, Ontario, Canada, M5G 2E9. * Corresponding author: E-mail: j.gagnie~toronto.ca
180
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
that these recommendations will lead to more complete and accurate reporting of herbal trials. We wrote this explanatory document to outline the rationale for each recommendation and to assist authors in using them by providing the CONSORT items and the associated elaboration, together with examples of good reporting and empirical evidence, where available, for each. These recommendations for the reporting of herbal medicinal products presented here are open to revision as more evidence accumulates and critical comments are collected. Key words : Herbal interventions, randomized controlled trials, CONSORT
INTRODUCTION Randomized controlled trials (RCTs) provide the best evidence for efficacy of health-care interventions (Altman et al., 2001). Carefully planned and well-executed RCTs give us the best estimates of treatment effect and can thusly guide clinical decision making (Mulrow et al., 1998; Sackett et al., 1998). Although, trials which lack methodological rigor cause over- or underestimations oftreatment effect sizes due to bias or confounding factors (Schulz et al., 1995; Moher et al., 1996; Jadad et al., 1996; Begg et al., 1996; Moher et al., 2001; The CONSORT Group). Hence, efforts have been undertaken towards improving the design and reporting of RCTs (Altman et al., 2001; Mulrow et al., 1998; Sackett et al., 1998; Schulz et al., 1995; Moher et al., 1996; J adad et al., 1996; Ioannidis et al., 2004; MacPherson et al., 2001). Current research suggests that reporting quality of complementary and alternative medicine (CAM) trials is poor (Linde et al., 2001a; Moher et al., 2002). Linde et al. (2001a) found that most CAM trials do not describe the generation of the random sequence, an adequate method of allocation concealment, and the number and reasons for drop outs and withdrawals. Moher et al. (2002) (The CONSORT Group, 2004) reported that a sample of pediatric CAM RCTs reported less than 40% of the CONSORT checklist items with a 24% increase in the number of checklist items included in reports over time. That is, less than half of all information necessary in the reporting ofthese trials appeared in their reports. Specifically, only 50% of trials reported how random numbers were generated and 25% if allocation concealment was done (Moher et al., 2002). The results suggest that a large proportion of CAM trials have poor reporting quality resulting in difficulties with assessment of internal and external validity (Linde et al., 200la; Moher et al., 2002). Linde et al. (2001a) showed that reporting quality may vary across different types of complementary therapies with herbal medicine 1 trials
CONSORT for Herbal Medicine Trials
181
being somewhat superior to homeopathy and acupuncture trials. Although, several systematic reviews state that trials of botanical medicine still fail to report information necessary to judge internal validity, external validity, and reproducibility (Gagnier et ai., 2003a; Gagnier, 2003b; Little & Parsons, 2000). A study examining the quality of reports of a sample of 206 English language herbal medicine RCTs found that less than 45% of the information suggested within the CONSORT statement was reported (Gagnier et ai., 2006a). For example, approximately 28% of trials described if the person administering the intervention was blinded to group assignment or not, only 22% described the methods for implementing the allocation sequence and 21% the method for generating the allocation sequence. Also, reporting quality differed between individual botanical medicines and improved across decades from the 1980s to the 2000s (Gagnier et ai., 2006a). Furthermore, it has been suggested that trials often do not include detailed information on the herbal product itself (Gagnier et ai., 2003a; Gagnier, 2003b). It is known that herbal medicines may vary by part of plant used, time of harvest, active constituent levels, type of extract (aqueous, alcoholic, glycerin) and delivery form. Therefore, results of clinical trials on heterogeneous products may vary considerably even if they are using the same botanical species. Variation in herbal products between trials precludes pooling in systematic reviews of herbal medicines since invalid inferences may result from the combined data (Gagnier et ai., 2003a; Gagnier, 2003b; Linde et ai., 2001b). It is clear that readers, editors and reviewers require increased transparency in the reporting of RCTs of botanical medicines. Reporting guidelines for controlled clinical trials have been developed.
The consolidated standards of reporting trials (CONSORT) statement was first published in 1996 and revised in 2001 (Begg et ai., 1996; Moher et ai., 2001). This statement consists ofa checklist and flow diagram to guide writers and reviewers on the information that should be available from published reports of two-group parallel randomized controlled trials (RCTs) (Begg et ai., 1996; Moher et ai., 2001). The CONSORT statement has been endorsed by many leading medical journals, editorial associations, professional societies and funding agencies (The CONSORT Group, 2004). Since its inception, several extensions of the CONSORT statement have been developed (Campbell et ai., 2004; Ioannidis et ai., 2004). Recently CONSORT was extended to cluster randomized trials (Campbell et ai., 2004) and for trials of harms Ooannidis et ai., 2004). Also, an international group of acupuncture researchers developed a set of recommendations for
182
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
improving reporting of the interventions in parallel group trials of acupuncture: the Standards for Reporting Interventions in Controlled Trials of Acupuncture or STRICTA (MacPherson et at., 2001). Though not a formal extension of CONSORT, MacPherson et at. (2001) describe STRICTA as a elaboration of item 4 in CONSORT and suggest STRICTA be used together with CONSORT in reporting acupuncture trials. In June 2004 an international group of trialists', methodologists, pharmacologists and pharmacognosists met for a consensus meeting in Toronto, Canada that led to the development of recommendations for the reporting of herbal medicine trials (Gagnier et at., 2006b; See Tables 1 & 2). The resulting guidelines amounted to a set of elaborations of current CONSORT items that will aid editors and reviewers in assessing the internal! external validity and reproducibility of herbal medicine trials, allowing an accurate assessment of safety and efficacy. During the development of the elaborations it became clear that an explanation of the concepts within and underlying the elaborations would aid researchers in planning, conducting and writing reports ofRCTs of herbal medicines. In the current paper we discuss the rationale and scientific background for each elaboration, provide examples of good reporting for each. Where possible, we discuss empirical evidence for each. It should be noted that each elaboration is an addition to existing CONSORT recommendations. Thus all CONSORT items are first listed in Table 1, and elaborations in Tables 2. Table 1. The CONSORT checklist (1,8) Standard Standard CONSORT CONSORT checklist: Paper checklist: Section and Topic Item Title & Abstract
Descriptor
Reported on Page number
How participants were allocated to interventions (e.g. "random allocation," "Randomized" or "randomly assigned"). Scientific background and explanation of the rationale.
Introduction Background
2
Methods Participants
3
Ehgibility criteria for participants and the settings and locations where the data were collected.
Interventions
4
Precise details of the interventions intended for each group and how and when they were actually administered.
Objectives
5
Specific objectives and hypotheses.
6
Clearly defined primary and secondary outcome measures and, when applicable, any methods used to enhance the quality of measurements (e.g. multiple observations, trainmg of assessors).
Outcomes
Table 1. Contd.
CONSORT for Herbal Medicine Trials
183
Table 1. Contd. Standard Standard CONSORT CONSORT checklist: Paper checklist: Section and Topic Item Sample size
7
Randomization Sequence allocation Allocation concealment
8
ImplementatIOn
10
Blindmg (Masking)
11
Statistical methods
12
Results Participant flow
13
Recruitment
14
Baseline data
15
Numbers analyzed
16
Outcomes Estimation
and
17
Ancillary analyses
18
9
Descriptor
Reported on Page number
How sample size was determined and, when applicable, explanation of any interim analyses and stopping rules. Method used to generate the random allocation sequence, includmg details of any restriction (e.g., blocking, stratification). Method used to Implement the random allocation sequence (e.g. numbered containers or central telephone), clarifying whether the sequence was concealed until interventIOns were assigned. Who generated the allocation sequence, who enrolled participants, and who assIgned participants to their groups. Whether or not participants, those administering the interventions, and those assessing the outcomes were blinded to group assIgnment. When relevant, how the success ofbhnding was evaluated. Statistical methods used to compare groups for primary outcome(s); Methods for additional analyses, such as subgroup analyses and adjusted analyses. Flow of participants through each stage (a dIagram is strongly recommended). Specifically, for each group report the numbers of participants randomly assIgned, receiving intended treatment, completing the study protocol, and analyzed for the primary outcome. Describe protocol deviations from study as planned, together WIth reasons. Dates defining the periods of recruItment and followup. Baseline demographic and clinical characterIstics of each group. Number of partiCIpants (denominator) in each group included in each analysis and whether the analysis was by "intention-to-treat". State the results in absolute numbers when feasible (e.g. 10120, not 50%). For each primary and secondary outcome, a summary of results for each group, and the estimated effect size and its precIsion (e.g 95% confidence interval). Address mUltiplicity by reportmg any other analyses performed, including subgroup analyses and adjusted analyses, indicating those pre-specified and those exploratory.
Table 1. Contd.
184
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Table 1. Contd. Descriptor
Standard Standard CONSORT CONSORT checklist: Paper checklist: Section and Topic Item
Reported on Page number
Adverse events
19
All important adverse events or side effects in each mtervention group.
Discussion Interpretation
20
Interpretation of results, taking into account study hypotheses, sources of potential bias or imprecision, and the dangers associated with multiplicity of analyses and outcomes.
Generalisability
21
Generalizabtlity (external validity) of trial results.
Overall evidence
22
General mterpretation of the results in the context of current evidence.
Table 2. Proposed elaborations of CONSORT items for randomized controlled trials of herbal medicine interventions*(20) Standard Standard CONSORT CONSORT checklist: Paper checklist: Section and Topic Item
Descriptor
Reported on Page number
How participants were allocated to interventions (e.g. "random allocation," "Randomized" or "randomly assigned"). Either the title or abstract. or both should state the herbal medicinal product s Latin binomial. the part of the plant lIsed. and the type ofpreparation.
Title & Abstract
Introduction Background
2
Scientific background and explanation of the rationale. Including a brief statement of reasons for the trial with reference to the specific herbal medicinal product being tested and. if applicable. whether new or traditional indications are being investigated
Methods Participants
3
Eligibility criteria for participants and the settings and locations where the data were collected. If a traditional indication is being tested. a description of how the traditional theories and concepts were maintained. For example. participant inclusion criteria should reflect the theories and concepts underlying the traditional indicatIOn.
Table 1. Contd.
185
CONSORT for Herbal Medicine Trials
Table 2. Contd. Standard Standard CONSORT CONSORT checklist: Paper checklist: Section and Topic Item InterventIOns
4
4.A. Herbal medicinal product name
Descriptor
Reported on Page number
Precise details of the interventions intended for each group and how and when they were actually administered. 1. The Latm binomial name together with botanical authority and family name for each herbal ingredient; common name(s) should also be included 2. The proprietary product name (i.e. brand name) or the extract name (e.g. EGb-76I) and the name of the manufacturer of the product 3 Whether the product used IS authorized (licensed. registered) in the country in which the study was conducted
1. The part(s) of plant used to produce the product or extract. 2. The type of product used [e.g raw (fresh or dry). extract] 3. The type and concentration of extraction solvent used (e.g., 80% ethanol, H20 IOO%, 90% glycerine etc.) and the herbal drug to extract ratio (drug:extract; e.g. 2'1) 4 The method of authentication of raw material (i e. how done and by whom) and the lot number of the raw material. State if a voucher specimen (I.e. retention sample) was retamed and, if so, where it is kept or deposited. and the reference number. 1. The dosage of the product, the duratIOn of 4.C. Dosage administration and how these were determined. regimen and 2. The content (e.g. as weight, concentration; may be quantitative given as range where appropriate) of all quantified descnption herbal product constituents, both native and added, per dosage unit form. Added materials, such as binders, fillers. and other excipients; e.g. 17% maltodextnn. 3% silicon dioxide per capsule. should also be listed) 3. For standardized products. the quantify of active/ marker constituents per dosage unit form 4.0. 1. Product's chemical fingerprint and methods used Qualitative (equipment and chemical reference standards) and who testing pe/formed it (e.g. the name of the laboratory used). Whether or not a sample of the product (i.e retention sample) was retamed and if so, where it is kept or deposited.
4.B. Characteristics of the herbal product
Table 1. Contd.
186
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation!
Table 2. Contd. Standard Standard CONSORT CONSORT checklist: Paper checklist: Section and Topic Item
Objectives Outcomes
4.E. Placebo! control group 4.F. Practitioner S 6
Sample size
7
Randomization Sequence allocation
8
Allocation concealment
9
Implementation
10
Blinding (Masking)
II
Statistical methods
12
Descriptor
Reported on Page number
2. Description of any special testing/purity testing (e.g. heavy metal or other contaminant testing) undertaken. Which unwanted components were removed and how (i.e. methods). 3. Standardization' what to (e.g. which chemical component(s) of the product) and how (e g. chemical processes, or biological/fimctional measures of activity) The rationale for the type of control/placebo used.
A description of the practitioners (e.g. training and practice experience) that are a part of the intervention. Specific objectives and hypotheses. Clearly defined primary and secondary outcome measures and, when applicable, any methods used to enhance the quality of measurements (e.g. multiple observations, training of assessors). Outcome measures should reflect the intervention and indications tested considering, where applicable, underlying theories and concepts.
How sample size was determined and, when applicable, explanation of any interim analyses and stopping rules. Method used to generate the random allocation sequence, including details of any restriction (e.g. blocking, stratification). Method used to implement the random allocation sequence (e.g. numbered containers or central telephone), clarifying whether the sequence was concealed until interventions were assigned. Who generated the allocation sequence, who enrolled participants, and who assigned participants to their groups. Whether or not participants, those admmistering the interventions, and those assessing the outcomes were blinded to group assignment. When relevant, how the success of blinding was evaluated. Statistical methods used to compare groups for primary outcome(s); Methods for additional analyses, such as subgroup analyses and adjusted analyses.
Table 1. Contd.
CONSORT for Herbal Medicine Trials
187
Table 2. Contd. Standard Standard CONSORT CONSORT checklist: Paper checklist: Section and Topic Item Results Participant flow
13
Recruitment
14
Baseline data
IS
Numbers analyzed
16
Outcomes Estimation
and
17
Ancillary analyses
18
Adverse events
19
Discussion Interpretation
20
Generalisabihty
21
Overall evidence
22
Descriptor
Reported on Page number
Flow of participants through each stage (a diagram is strongly recommended). Specifically, for each group report the numbers of participants randomly assigned, receiving intended treatment, completing the study protocol, and analyzed for the pnmary outcome. Describe protocol deviations from study as planned, together with reasons. Dates defining the periods of recruitment and followup. Baseline demographic and clinical characteristics of each group.lncluding concomitant medication, herbal and complementary medicine use. Number of participants (denommator) in each group included in each analysis and whether the analysis was by "intention-to-treat". State the results in absolute numbers when feasible (e.g. 10/20, not 50%). For each primary and secondary outcome, a summary of results for each group, and the estimated effect size and its precision (e.g. 95% confidence mterval). Address multiplicity by reportmg any other analyses performed, including subgroup analyses and adjusted analyses, indicatmg those pre-specified and those exploratory. All important adverse events or side effects in each intervention group. Interpretation of results, taking into account study hypotheses, sources of potential bias or imprecision, and the dangers associated with multiplicity of analyses and outcomes. Interpretation of the results in light of the product and dosage regimen used. Generalizability (external validity) oftrial results. Where possible, discuss how the herbal product and dosage regimen used relate to what is used in self-care and/or practice. General interpretation of the results in the context of current evidence. Discussion of the trial results in relation to trials of other available products.
*CONSORT items (1,17) are listed In normal text. Proposed recommendations for reports of herbal medic me RCTs are listed in italicized text.
188
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
CONSORT ELABORATIONS INTERVENTIONS
FOR TRIALS
OF
HERBAL
Where necessary, we have elaborated items, to describe information suggested in reports of RCTs of herbal medicinal product interventions. When reporting an RCT of an herbal medicine, authors should consider reporting the information outlined in the CONSORT statement in addition to the information suggested in these elaborations. Below we list original CONSORT items (in normal type) followed by elaborations (in italicized text) for RCTs of herbal interventions. Excerpts from trials include information suggested by the CONSORT items and the developed elaborations.
TITLE AND ABSTRACT Item 1. How participants were allocated to interventions (e.g. "random allocation," "Randomized" or "randomly assigned"). Either the title or abstract, or both should state the herbal medicinal product's Latin binomial, the part of the plant used, and the type of preparation. Examples Title: "A double-blind, placebo-controlled, randomized trial of Ginkgo biloba extract EGb 761 in a sample of cognitively intact older adults: neuropsychological findings" (Mix & Crews, 2002) Abstract: "This was a randomized, double-blind placebo controlled ... The active treatment group received tablets containing 300 mg of Garlic Powder (Kwai) ..... This is equivalent to approximately 2.7 g or approximately 1 clove of fresh Garlic per day." (Isaacshon et al., 1998).
Explanation CONSORT item #1 is meant to aid in the indexing and identification of reports of randomized controlled trials (RCT) using electronic databases (Altman et al., 2001). Hence the use of the words "randomized", "randomly" or "random allocation" is suggested in the CONSORT statement. Additional language is required in the title and abstracts of trials of herbal medicinal products. The practice of evidence-based herbal medicine requires access to the herbal scientific literature. The identification of RCTs of herbal medicinal products requires that the product's Latin binomial, the part of the plant used and the type of preparation be reported in the title and/or abstract. This information would allow increased specificity in the indexing and identification of RCTs of particular herbal medicine products or preparations. Some herbal medicinal products have a specific trade name or commercial name. Where applicable, this name should be listed, together with the Latin binomial of the ingredient herb. When the herbal medicinal product used in the trial is a combination herbal product, we suggest listing
CONSORT for Herbal Medlcine Trials
189
the product name in the title and the separate herbal medicinal species contained within this product in the abstract. In this way, the title of the trial will not be prohibitively long by listing all separate herbal species' Latin binomials. Studies indicate that searching for complementary and alternative medicine (CAM) related topics is challenging due to diversity of use of controlled vocabulary and indexing procedures between different databases (Murphy et al., 2003). It has been suggested that if authors of CAM trials (e.g. botanical medicine trials) report abstracts or titles without reference to standard controlled vocabulary, indexers may not assign appropriate indexing terms for particular studies (Murphy et al., 2003; McGregor, 2002). A further problem arises from indexers not having a sufficient number and variety of descriptors for CAM interventions (Kronenberg et al., 2001). When reporting RCTs of herbal medicinal products, the use of the information suggested for titles and abstracts above will likely lead to improved indexing and retrieval.
INTRODUCTION Background
Item 2. Scientific background and explanation of the rationale. Including a brief statement of reasons for the trial with reference to the specific herbal medicinal product being used, and if applicable, whether new or traditional indications are being tested. ExampleA The extract of Ginkgo biloba leaves entitled EGb 761 is a complex mixture that is standardized with respect to its flavonol glycoside (24%) and terpene lactone (6%) content [1,2]. These two classes of compounds have been implicated in the benefcial effects of EGb 761 in treating peripheral and cerebral vascular insuffciency, age-associated cerebral impairment, and hypoxic or ischemic syndromes [1,3]. Electron spin resonance (ESR) studies conducted in vitro have shown that EGb 761 is an efficient scavenger of various reactive oxygen species, including superoxide anion radical (0282) and hydroxyl radical (H08), and that it also exhibits superoxide dismutase-like activity [4]. Recent in vitro studies with animal models have revealed that the extract may exert an anti-free radical action in myocardial ischemia-reperfusion injury. In these studies [5,6], inclusion of 200 mg/l of EGb 761 in the medium that was used to perfuse isolated ischemic rat hearts signifcantly improved postischemic recovery, reduced ventricular arrhythmias and enzyme leakage, and lowered the content of A Please
note, all references listed as numbers in all quotes provided refer to the reference number in the original publication from which this excerpt was taken.
190
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
spin-trapped oxyradicals in the coronary effluents. Interestingly, antiarrhythmic effects were also observed when animals were treated orally with EGb 761 prior to heart perfusion, but a significant reduction in ventricular arrhythmias could be achieved only with high dosages (.100 mg/kg for 10 days) [5]. In addition to these studies conducted with EGb 761 [5,6], numerous other studies with experimental animals have indicated that active reduced forms of molecular oxygen, including 0282, HOS, and hydrogen peroxide (H202), are involved in the pathogenesis of tissue injury that follows myocardial ischemia-reperfusion [7-10] (Pietri et al., 1997) ........................ In the present double-blind study, we tested the cardioprotective efficacy of oral treatment with EGb 761, which is known to have in vitro antioxidant properties [4-6], in patients undergoing CPB surgery by manual palpation (Pietri et al., 1997).
Explanation The background of reports of controlled clinical trials partially serve to layout the rationale of the trial (Altman et al., 2001) with special reference to the specific intervention under study. There is great heterogeneity in the types of herbal medicinal products available. Two different preparations/ products of the same herbal species can have different phytochemical profiles, differing pharmacokinetic properties, etc. Given the variability in products, the rationale should clearly overview the scientific data concerning the specific herbal medicinal product under study (e.g. in the above example EGb 761). Where no clinical trials are available for review, extrapolation from pre-clinical work (i.e. animal studies, observational studies, case reports, known mechanisms) is acceptable. Where no data on the product is available, previous research on similar products to that being tested in the current trial should be reviewed. This information should be clearly stated and ideally include a description of a systematic review of previous studies utilizing the herbal product (Altman et al., 2001; Savulescu et al., 1996). Also, if the authors are testing a traditional use, a review of the theory and concepts underlying this indication should be reported. Readers with some relevant knowledge of the area should be able to determine the reasoning for the indication. For example, trials of traditional Chinese medicine (TCM) may choose to test a TCM diagnosis (e.g. Liver Blood Deficiency) and not a western diagnosis (Hepatitis). If this is so, the authors should be explicit in their description of why the particular intervention being tested is indicated. For example,
In traditional Chinese medicine, the "Nei-Kuan" acupoint (EH-6, where EH denotes equilibrium envelope of the heart meridian) has been believed to correlate with the function of the heart (Chuang, 1977). Recently, some investigators (Mah et al., 1992; Hsu et al., 1989) observed that
CONSORT for Herbal Medicine Trials
191
acupuncture at Nei-Kuan can improve left ventricular function in patients with coronary heart disease (Ho et aI., 1999). Additionally, other data (i.e. clinical trials, animal studies, observational studies, case reports, known/proposed mechanisms) that would aid in creating a rationale, even for this traditional indication, can be described in the background. The assumption is that a rationale can be clearly and explicitly reported and that it may be derived from scientific, empirical, historica1 4 or traditiona1 5 sources.
METHODS Participants
Item 3. Eligibility criteria for participants and the settings and locations where the data were collected. If a traditional indication is being tested, a description of how the traditional theories and concepts were maintained. For example, participant inclusion criteria should reflect the theories and concepts underlying the traditional indication.
Example There were altogether 118 cases, which were randomly divided into two groups: The QTG (Qingluo Tongbi Granules: A Chinese herbal medicine) treated group and the control group treated with tripteryguim glycosides. In the treated group (n=63), there were 18 males and 45 females, aged from 18 to 65 years with an average of 39.5 ± 16.6 and the disease course ranging 2-22 years averaging 7.5 ± 3.6 years. The cases were graded by xray according to the criteria set by the American Association of Rheumatoid Arthritis (ARA), USA: 7cases were grade I, 30 grade II, and 26 grade III. In the control group (n=55), there were 10 males and 45 females, aged from 18 to 65 years with an average of 38.3 ± 16.7 and the disease course ranging 1-21 years, averaging 6.9 ± 3.1 years. Among them, 15 cases were grade I, 21 grade II, and 19 grade III. The cases in the two groups were comparable in sex, age, disease course and X-ray grading (p>0.05). Diagnostic criteria and TCM differentiation criteria: Diagnosis of RA was made according to the ARA criteria revised in 1987. Criteria for TCM differentiation of the type of yin-deficiency and heat in collaterals: burning pain in joints, local swelling, or deformity and rigidity, reddened skin with a hot sensation, low fever, dry mouth, yellow urine, red or dark red tongue with ecchymosis and petechia, thin or yellow and greasy or scanty fur with fissures, fine, rapid and slippery or fine and rapid pulse. Included in the study were inpatients and outpatients who were diagnosed to have RA of the type of yin-deficiency and heat in the collaterals (Italicized words added from a previous passage in the manuscript) (Xueping et al., 2004).
192
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Explanation The external validity, generalizability, of a trial is partially dependent on the eligibility criteria for participants (Altman et al., 2001). Reporting of eligibility criteria in trials of herbal medicine interventions is often poor. One study found that less than 75% of RCTs of herbal interventions adequately reported eligibility criteria (Gagnier et al., 2006a). As a result, determination of generalizability of one quarter of these trials would not be possible from reading the published reports. Trials of herbal medicines that aim to test traditional indications must be sure to report any eligibility criteria which reflect this. On a related note, authors may choose to exclude participants with previous use of the specific herbal medicinal product itself. It has been suggested that use of herbal medicinal products prior to trial commencement can lead to increased amounts of adverse effects. A trial of Tanacetum parthenium (Feverfew) (Johnson, 1984) against placebo for migraine prophylaxis included current use of feverfew as an eligibility criterion. Those in the placebo group experienced more side effects, these were attributed to a "post-feverfew syndrome" which is the equivalent of withdrawal effects. Such symptomatic worsening following cessation oflongterm feverfew consumption has been reported elsewhere (Johnson, 1984). To date there is no empirical evidence to suggest that use of an herbal medicine prior to a controlled clinical trial of that same herbal medication biases estimates of treatment effect. Although, the use and reporting of eligibility criteria to exclude trial participants with recent use is suggested.
Example Anyone with a prior adequate trial of St John's wort (at least 450 mg/d) for the treatment of depression or those who had taken St John's wort for any reason in the last month were excluded. To reduce the potential for including a treatment nonresponsive sample, participants who had failed to respond to a trial of an antidepressant (fluoxetine hydrochloride, 20 mg/d, for at least 4 weeks or the equivalent) in the current episode or who had failed to respond to more than 1 adequate trial of antidepressant in a previous episode also were excluded (Shelton et al., 2001).
Explanation It is important that trials of herbal medicines report the settings and locations where the data where collected (Altman et al., 2001). The location highlights physical factors (e.g. climate, food sources), economics, geography and social and cultural factors that may affect the generalizability of a study. As well, research settings may vary greatly in their organization, resources, experience, baselines risk, and physical appearances (Altman et al., 2001). One study found that less than 40% of herbal medicine trial reports adequately reported the setting and location of the trial (Gagnier
CONSORT for Herbal Medicine Trials
193
et al., 2006a). External generalizability of trial results partially rests on complete reporting of this information. Item 4: Precise details of the interventions intended for each group and how and when they were actually administered. Where applicable, the description of a herbal intervention should include: 4.A.
Herbal medicinal product name
1. The Latin binomial name together with botanical authority and family name for each herbal ingredient; common name(s) should also be included 2. The proprietary product name (i.e. brand name) or the extract name (e.g. EGb-761) and the name of the manufacturer of the product 3. Whether the product used is authorized (licensed, registered) in the country in which the study was conducted
4.B. Characteristics of the herbal product
1. The part(s) of plant used to produce the product or extract. 2. The type of product used [e.g. raw (fresh or dry), extract] 3. The type and concentration of extraction solvent used (e.g. 80% ethanol, H 2 0 100%, 90% glycerine etc.) and the herbal drug to extract ratio (drug:extract; e.g. 2:1) 4. The method of authentication of raw material (i.e. how done and by whom) and the lot number of the raw material. State if a voucher specimen (i.e. retention sample) was retained and, if so, where it is kept or deposited, and the reference number.
4.C. Dosage regimen and quantitative description
1. The dosage of the product, the duration of administration and how these were determined. 2. The content (e.g. as weight, concentration; may be given as range where appropriate) of all quantified herbal product constituents, both native and added, per dosage unit form. Added materials, such as binders, fillers, and other excipients; e.g. 17% maltodextrin, 3% silicon dioxide per capsule, should also be listed) 3. For standardized products, the quantity of active / marker constituents per dosage unit form
4.D. Qualitative testing
1. Product's chemical fingerprint and methods used (equipment and chemical reference standards) and who performed it (e.g. the name of the laboratory used). Whether or not a sample of the product (i.e. retention sample) was retained and if so, where it is kept or deposited. 2. Description of any special testing/purity testing (e.g. heavy metal or other contaminant testing) undertaken. Which unwanted components were removed and how (i.e. methods). 3. Standardization: what to (e.g. which chemical component(s) of the product) and how (e.g. chemical processes, or biological/ functional measures of activity)
4.E. Placebo/control group
The rationale for the type of control/placebo used.
4.F.
A description of the practitioners (e.g., training and practice experience) that are a part of the intervention.
Practitioner
194
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
The type of information that is required for a complete description of any intervention is relative to the type of intervention being tested. For trials of surgical interventions, for example, a complete description of the individual performing the surgery may be required (Altman et al., 2001; Roberts, 1999). For herbal medicines, the above information is required to determine, with specificity, the key characteristics of the product that was used. A complete description of the product will allow determination of its' efficacy and safety relative to other products. There are a wide variety of commercially available products containing herbal medicines (Dreikorn et al., 1998; Dreikorn et al., 2001; Harkey et al., 2002). In addition there is great variability in the content of these products (Harkey et al., 2002; Gurley et al., 2000; Liberti & Der Marderosian, 1978; Groenewegen & Heptinstall 1986; Heptinstall et al., 1992; Nelson et al., 2002; Zhang et al., 2002). Often products do not contain the amount (weight, volume, proportion) of the individual constituents listed on their label (Manning & Roberts, 2003) or any of the constituents at all. Products containing the same botanical species [e.g. Hypericum perforatum (St. John's wort)] often contain varying amounts of the plant's marker/active 6 constituents. For example, research has shown that commercial products of the following botanical species contain varying levels of their respective constituents: Hypericum perforatum (St. John's wort) (Garrara et al., 2003; Schulte-Lobbert et al., 2004), Camellia sinensis (Green tea) (Manning & Roberts, 2003), Tanacetum parthenium (Feverfew) (Gronewegen and Heptinstall, 1986; Heptinstall et al., 1992; Nelson et al., 2002), Eleutherococcus senticosis (Siberian ginseng), Panax quinquefolius (American/Canadian ginseng) (Harkey et al., 2002), Hydrastis canadensis (Goldenseal) (Weber et al., 2003), and Paullinia cupana (Guarana) (Carlson & Thompson, 1998). As a result, the pharmacological properties and in vitro activities may vary between different products (e.g. Dreikorn et al., 2001; Lefebvre et al., 2004). Also, some studies have shown that botanical products not only contain varying beneficial constituents, but also varying harmful ones (Haller et al., 2004; Oberlies et al., 2004). Therefore, it is necessary that authors of trials of herbal interventions completely describe the product used.
Herbal medicinal product name Example The AG (American Ginseng) capsules contained 3-y-old Ontario grown, dried, ground AG root (Panax quinqueefolius L.) supplied by the same supplier, Chai-Na-Ta Corp., BC, Canada, ..... This AG was the same commercially available product, but from a different batch than the original (Sievenpiper et al., 2003).
CONSORT for Herbal Medicine Trials
195
Explanation
Reports should state the Latin binomial and common name/names together with the authority and family name. Reporting of the Latin binomial and common name were also suggested in item 1. The accepted international code of botanical nomenclature (Greuter et al., 1999) indicates that the scientific naming of botanical species must include a Latin binomial (genus and specific epithet) and the authority name. For example, Genus: Taraxicum, Epithet: officinale Authority: Linnaeus. In full, this would result in: Taraxicum officinale L. (Linnaeus is abbreviated as L. here). The authority identifies who originally described the plant. Common names should also be listed here (e.g. Dandelion, Feverfew, St. John's wort). Alone, common names are not sufficient since different herbal species may have the same common name. For example, Echinacea is a common name used for Echinacea angustifolia, Echinacea pallida, and Echinacea purpura. These plants have heterogeneous biochemical profiles (Pellati et al., 2004). If relevant, the proprietary product7 name (i.e. brand name, e.g. Kwai) or the extractS name (e.g. LI 160) and the manufacturer of the product should be reported. Such names are a quick means of identification of the specific herbal product including its contents and manufacturing or production. Alone, these names are not sufficient for the product description. Authors should also report whether the product is licensed in the region where the trial took place. Specific regulatory bodies award licenses for herbal medicinal products. The regulations for attaining licensing are variable across jurisdictions. Though licensing does not ensure product quality or provide the reader with a sufficient description of the herbal product, it does allow the reader to determine the regulatory status and availability of a specific herbal medicine.
Physical characteristics of the herbal product Example (raw herb)
The ginseng capsules contained 3-y-old Ontario dried and ground ginseng root (P. quinquefolius L.) ...... All ginseng and placebo capsules came from the same lot (Vuksan et al., 2001). Example (extract)
... all patients received 1 infusion/day with Ginkgo special extract Egb 761 (batch number: 5242) over 30 - 60 min (1 dry vial in 500 mL isotonic solution). The dry vials contained 200 mg of dry extract from Ginkgo biloba leaves (drug-extract ratio 50 : 1), .... :12 H 20 in 3 mL solution served as solvent (Morgenstern & Biermann, 2003). Explanation
There must be a complete description of the physical characteristics of the herbal product including the partes) of the plant contained in the product or
196
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
extract and the type of product [e.g. raw (fresh or dried) or extract]. The partes) of the plant included in a product are related to the quantities and types of constituents present (Betz et al., 1994). Also, the type of product used should be reported given that different product forms have different types and amounts of constituents (Patora et al., 2003). If the product is an extract, the type and concentration of the extraction solvent should be reported (e.g. 80% Alcohol, H 20 100%, 90% glycerine etc.) as well as the plant to plant extract ratio (plant: plant extract; e.g. 2:1). This ratio tells the reader how much of the starting plant material (either by weight or volume) was required to produce a specific amount ofthe finished extract. The method of authentication of raw material (i.e. how done and by whom) describes how the original material or plant was identified and allows the reader to determine, to some degree, if the raw material for the herbal product was produced from the plant as reported. The lot number of the raw material provides the reader with key information as to where the raw material came from.
Dosage regimen and quantitative description
Example The treatment was provided as 252 tablets containing 50 mg of either Ginkgo biloba standardized extract LI 1370 (containing 25% flavanoids, 3% ginkgolides, and 5% bilobalides) or placebo (both provided by Lichtwer Pharma). Participants were instructed to take three tablets daily for 12 weeks. The extract and dose of Ginkgo biloba were chosen on the basis of the results of previous trials in which this dose of this extract had been reported to be effective in treating cerebral insufficiency> (Drew & Davies, 2001).
Explanation Authors of trials of herbal medicines should report the dosage regimen and provide a quantitative description of the herbal product. Information regarding the dosage and duration of the trial are of great importance to replicating trials, establishing efficacy or harm for specific dosages and durations, and for external generalizibility (Altman et al., 2001). The rationale for dosage and duration of the trial should be clear as unclear reasoning questions the methods of a trial and possibly raises some ethical issues as to why the trial was carried out at all. The weight or amount of all herbal product constituents, both native and added per dosage unit form (i.e. added materials such as binders, fillers, excipients, etc; e.g. 17% maltodextrin, 3% silicium dioxide per capsule) and the percentage of active/marker constituents per dosage unit form (e.g. 0.3% Hypericin per capsule) should also be reported. This provides the reader with a profile of the quantity of the botanical product constituents.
197
CONSORT for Herbal Medicine Trials
Qualitative testing Example The content of various ginsenosides (RgI' Re, Rf, Rb, Rc, Rb 2 , and Rd), which are dammarane saponin molecules found among Panax species, was determined in the laboratory of Dr. John T. Arnason at the Department of Biology, Faculty of Science, University of Ottawa, Ontario, Canada, using high-performance liquid chromatography (HPLC) analyses, a method similar to the one developed for the American Botanical Council Ginseng Evaluation Program [27]. A Bechham HPLC system with a reverse-phase Beckham ultrasphere C-18, 5um octadecylsilane, 250 X 4.6mm column was employed for the analyses. The ginsenoside standards used for comparison were provided by two sources. Rg 1 and Re were provided by Dr. H. Fong, University of Illinois and Rf, RbI' Rc, Rb 2 , Rd were provided by Indofine Chemical Co., Somerville N.J. (Vuksan et al., 2000) Table 1. Energy. nutrient, and ginsenodide profile of the American ginseng (Panax quinquefolius L.) and placebo capsules Constituent (per g) E nergy 2 (kJ) 14.68 (kcal) Macronutrients 2 Carbohydrate (g) Fat (g) Protein (g) Ginsenosides (20S)- Protopanaxadiols (%) RbI Rb2
Rc
Rd (20S)-Protopanaxatriols (%) Rgi Re Rf Total (%)
Placebot
Ginsengt
14.39 3.51
3.44
0.73 0.039 0.069
0.57 0.013 0.26
1.53 0.06 0.24 0.44 0.100 0.83
o
3.21
ITo equate energy and macronutrient values to 1, 2, or 3 g American ginseng, multiply by 1, 2, or 3, respectively. To determine values for placebo, multiply by 2. 2Determined by the Association of Official Analytical Chemists methods for macrountrients (18). 3Determined by HPLC analyses (20). Adopted from: Vuksan et al., 200l.
Explanation Trials should report the product's chemical fingerprint 9 and methods used (machinery and chemical reference standards) and who performed it (the
198
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
name ofthe laboratory used). The fingerprint can be reported in a graph or a table describing the key constituents of the herbal medical product. Chemical profiling, using the proper techniques is essential to providing a clear and accurate report of a product's constituents, and provides both qualitative and quantitative information (Sawnson, 2002; Phytomedicine: International Journal of Phytotherapy and Phytopharmacology; German Federal Institute for Drugs and Medical Device, 1999). Bauer and Tittel (1996) have provided some guidelines for the characterization and standardization of plant material used for pharmacological, clinical and toxicological studies. Also the Association of Analytical Communities (AOAC) has outlined standards for analyzing specific herbal medicinal products (AOAC, 2004) as has the American Herbal Pharmacopoeia produced by the American Botanical Council (American Herbal Pharmacopoeia, 2004) and the United States Pharmacopoeia (United states Pharmacopoeia, 2004). Reports might also describe if a voucher specimen 10 (i.e. retention sample) was retained and if so, where it is kept or deposited, so that independent sources can verify the chemical profile. Herbal medicines are often contaminated (Chan, 2003). Thus, a complete description of any special testing/purity testing (e.g. heavy metal or other contaminant testing) and the removal of unwanted components (which and how (the methods» should be included in reports where relevant. All such methods are relevant given that they may alter the composition of the herbal product. Standardization has been hotly debated in the literature (e.g. Swanson, 2002). Often, companies or researchers attempt to standardize botanical products to specific chemical "marker" constituents. These "marker" constituents may be considered to be the primary "active" constituents or merely serve an index of the product's chemical profile (Swanson, 2002). Though products on the commercial market may not be standardized and often an "active" constituent is not known, if standardization was done in a clinical trial of an herbal product, it should be reported. Authors should report, what the product was standardized to (e.g. which chemical component(s», how this was done (i.e. chemical processes, or biological! functional measures l l of activity) (e.g. Murphy et al., 1988) and the percentage of this particular constituent per dosage unit form. The rationale for the type of control/placebo used.
Example The placebo, on the other hand, consisted of identical capsules containing corn flour. The energy, carbohydrate content and appearance ofthe placebo were designed to match that of the AG (American Ginseng capsules). (Italicized words added). (Sievenpiper et al., 2003)
CONSORT for Herbal Medicine Trials
199
Explanation In botanical medicine trials, as in other trials, it is important to have a complete description ofthe characteristics of the control group and the way in which it was disguised (Altman et al., 2001). If a placebo control was used, the placebo should be closely matched to the control intervention (Vickers & de Craen, 2000). For trials of herbal interventions the rationale for the type of control/placebo used should be described. There have been some trials that have reported using placebos that are matched to color and smell, but that contain ingredients that are active (e.g. Palevitch et al., 1997). If a control group is active, comparisons between it and the experimental group will be affected. While it may be a challenge to construct matched placebos for certain herbal product interventions, it is not impossible. A description of the practitioners that are a part of the intervention.
Example In order to participate in the study, physicians had to (i) be a medical specialist with a degree in internal medicine and general medicine, (ii) have a certified degree in TCM by a German society for medical acupuncture, and (iii) have at least 5 years of practical experience in TCM (according to the German Acupuncture Societies Working Group standard) ....... The herbal formulations for the TCM group were designed by a herbalist (CarlHermann Hempen) and prepared by a pharmacist who both specialise in Chinese herbal medicine (S. Dietz, Franz-Joseph-Pharmacy, Munich, Germany). In addition to the basic formula, every patient received a second additional formula tailored to his or her individual TCM diagnosis (Table 1). (Brinkhaus et al., 2004)
Explanation On occasion an herbal intervention trial may include a health-care practitioner as part of the intervention. Practitioners have varying levels of training, years of experience, theoretical orientations and work in different environments. Similar to surgical trials, such trials should provide a description of the practitioners (e.g. training and practice experience) that are a part of the intervention (Altman et al., 2001).
Outcomes Item 6: Clearly defined primary and secondary outcome measures and, when applicable, any methods used to enhance the quality of measurements (e.g., multiple observations, training of assessors). Outcome measures should reflect the intervention and indications tested considering, where applicable, underlying theories and concepts.
200
Comp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Example All outcome measures were assessed at baseline and after 30 days of treatment at the follow-up visit. The primary outcome measures were changes in quality oflife as measured by the Physical and Mental Component Summary scales of the 12-Item Short Form Health Survey (SF-12). The SF-12 is widely used in measuring health and quality of life and has been shown to have a high level of agreement with scores from the original 36Item Short Form Health Survey (SF-36)[11]. The SF-36 has been validated in several Chinese studies, whereas evaluation of the SF-12 is ongoing [11]. Secondary outcome measures included assessments of physical performance, memory, sexual function, and qi ................. The qi scale is a 17-item instrument (14 items on an interviewer-administered questionnaire and three physical examination items) that was developed through an international collaboration of clinical investigators with expertise in scale development and traditional Chinese medicine. Questionnaire items address symptoms commonly included in a traditional Chinese medical interview, including breathing, energy level, appetite, heartburn, sweating, bowel patterns, pain, temperature sensations, sleep habits, and sexual ability. The physical examination items address tongue coating, tongue muscle quality, and pulse quality. The scale was developed for this study and has not been validated. The 14 questionnaire items are scored on a scale of 0 to 4 points, and the physical examination items are scored on a scale of 0 to 3. The total qi score is the sum of each score, ranging from 0 (best) to 65 (worst) (Bent et at., 2003). Explanation As with any RCT, outcome measures, both primary and secondary, should be relevant to the indications being tested be fully reported and describe any methods used to enhance the quality of measurements (1,8)(Altman et al., 2001; Moher et al., 2001). When performing RCTs testing herbal interventions, concepts that go beyond western medical terminology and understanding may be relevant. For example, in the above trial the particular Chinese herbal remedy being tested is purported to increase longevity, quality oflife, energy, memory, sexual function and Qi, a Chinese concept that is loosely translated as vital energy. Therefore, in addition to measures of health and quality of life, these investigators required a measure of Qi to test the change in vital energy during the course of this trial. Ultimately, to test the function of traditional herbal medicines we advise that the outcome measures reflect the underlying theories and concepts and therefore the indications for the specific herbal medicine intervention under investigation.
CONSORT for Herbal Medicine Trials
201
RESULTS Baseline data
Item 15: Baseline demographic and clinical characteristics of each group. Including, concomitant medication use or herbal medicinal product use. Example Eight patients (mean age 44.9 (SEM 4.2) years) received feverfew and nine (mean age 51.2 (2.3) years) received placebo capsules. The patients in the active group had taken 2.44 (0.2) small leaves of feverfew daily for 3.38 (0.58) years before entry to the study, and those in the placebo group had taken 2.33 (0.48) small leaves daily for 4.18 (0.67) years. Thus the two groups did not differ in the amount of feverfew consumed daily or the duration of consumption ..... . One patient in each group was taking conjugated equine estrogens (Premarin); the patient in the placebo group was also taking pizotifen. One patient given feverfew was taking the combined oral contraceptive Orlest 21. On patient in each group was taking a diuretic: the patient given feverfew was taking clorazepate and the patient given placebo was also taking a product containing tranylcypromine and trifluorperazine. In addition, two people in the placebo group were taking vitamin preparations and one prochlorperazine (Johnson et al., 1985).
Explanation A complete description of participants who entered a trial allows readers and clinician's to assess how relevant the trial is to a specific patient. As a part of the baseline assessments in trials of herbal medicinal products, authors should clearly assess and describe any current medication or herbal product use. Differences between groups on medication or herbal product use may confound results (Piscitelli & Burstein, 2002).
DISCUSSION Interpretation
Item 20: Interpretation of results, taking into account study hypotheses, sources of potential bias or imprecision, and the dangers associated with multiplicity of analyses and outcomes. Interpretation of the results in light of the product and dosage regimen used. Example Although EGb 761 is generally used at a dose of 120 mg/day in treating chronic disease states, we chose to administer the extract at more than twice its usual dose, but for only 5 days before the operation, to cope with the enhanced generation of oxidant species that was expected to follow
202
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
post-unclamping procedures. Measurements of DMSO/AFR concentrations indicated that EGb 761 treatment significantly protected plasma ascorbate levels in all sampling sites during the initial 5-10 min. ofreperfusion (see Table 3), a period during which free radical processes are considered to be critical. Analyses of plasma TBArs concentrations revealed that EGb 761 treatment also suppressed (or substantially attenuated) the transcardiac release of MDA, indicating protection against free radical-induced lipid peroxidation. These two findings offer some clues regarding the mechanisms that underlie the protective action of EGb 761 in open-heart surgery. It has been reported that EGb 761 protects ischemic rat hearts against reperfusion-induced decreases in ascorbate [6]. We did not observe any in vitro chemical interaction between EGb 761 and AFR or DMSO/AFR that would have maintained ascorbate levels. Therefore, in performing its antioxidant role, it seems that the extract competes effectively with circulating ascorbate (e.g. by directly quenching oxyradicals once they have been formed) ................ (Pietri et al., 1997).
Explanation The discussion section of a clinical trial report provides the reader with information to determine the pros and cons of the study and how this study relates to other research in the area. Understanding how the results with this product and dosage regimen relate to the results of other trials with similar or different products or dosage regimens allows the reader to establish a context from which to determine strengths or drawbacks of the specific intervention used. Therefore, authors should clearly state specific aspects of the product or dosage regimen that could have resulted in trial findings. This concept is discussed here under Item 20, to highlight the necessity of authors of reports of RCTs of herbal products to explicitly consider the product and dosage regimen as potential strengths or drawbacks of the study. Though we have formally separated this from the elaboration on Item 22, discussion of the trial results in the context of the current evidence (Item 22), we admit that these aspects of the discussion section are closely related, and may be written together in the body of the manuscript.
Generalizability Item 21: Generalizability (external validity) of trial results. Where possible, discuss how the herbal product used relates to what is used in self-care and I or practice. Example G 115 is a standardized ginseng extract which is often complexed with various other substances and marketed commercially. Ginsana®, Gericomplex®, Geriatric Pharaton®, and ARM229 are several commercial standardized
CONSORT for Herbal Medicine Trials
203
ginseng products which have been studied, and may include some or all of the following substances in addition to G 115: vitamins, minerals, trace elements, dimethylaminoethanol bitarate. A word of caution for the consumer. As noted previously, the FDA classifies ginseng as a food supplement, so it is marketed rather extensively in health food stores. An estimated 5-6 million Americans use ginseng products (6). However Chong and Oberholzer (11) note that there are problems with quality control, and indeed a recent report (12) indicated that of 50 commercial Ginseng preparations assayed, 44 contained concentrations of ginsenosides ranging from 1.9% to 9.0%, while six of the products had no detectable ginsenosides (Dowling et al., 1996).
Explanation Generalizability, or external validity is the extent to which the results of a study hold true in other circumstances (Fletcher et al., 1996). The word "circumstances" here can mean other individuals or groups of individuals, other similar interventions, dosages, timing, administration routes, and other settings for starters. Given the wide variability in herbal medicinal products available on the market, and their variable quality and content, a review of how the products used in the current trial relate to what is available and/or used by consumers and practitioners is quite valuable. This information would allow the reader to determine the availability of products that may act similarly to the one used in the triaL Application of clinical trial results partly relies on the availability of the intervention or a similar intervention.
Item 22: General interpretation of the results in the context of current evidence. General discussion of the trial results in relation to trials of other available products. Example The majority of published studies to date have used a powdered garlic preparation, similar to the preparation method used in this study. Considerable variability in outcomes exists between these studies. For example, Adler et al. [13], using a commercial dehydrated garlic tablet, reported a significant net drop of 13.1% in LDL-C levels relative to the placebo group in 12 weeks, and Jain et al. [15], using the same product and a similar design, reported a significant net decrease of 8% in LDL-C levels in moderately hypercholesterolemic adults. However, three other studies [19,20,22], using the same dosage of the same commercial dehydrated garlic powder product (Kwai®, Lichtwer Pharmaceuticals) reported no significant effect. The dose of powdered garlic tablets used in the five studies just cited, 900 mg:day, was similar to the full dose of 1000 mg:day used in this study. The allicin content of the tablets used in this study, 1500 mg:day in the full dose, was lower than the amount used in other studies with powdered
204
Camp. Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
garlic preparations. Other types of garlic preparations used in lipid lowering trials have included aged garlic extract and teamed garlic oil. Steiner et al. [14] used a large dose, 9 tablets: day, of aged-garlic extract, and reported a statistically significant 4.6% lowering of plasma LDL-C levels. In contrast, a recent study using steamed garlic oil supplementation reported no significant effect on cholesterol levels in hypercholesterolemic adults after 12 weeks [18]. One explanation could be that the oil is not as effective as dehydrated garlic powder because it contains different sulfur-containing phytochemicals [30]. Some of the discrepancies reported in these studies can be explained by the heterogeneity that exists among them in terms of study design, duration, subject characteristics, adherence, or confounders such as weight, diet, and exercise (Gardner et al., 2001).
Explanation Discussing trial results in the context of relevant studies is important to put the trial results in the context of existing empirical evidence (Altman et al., 2001; Annals ofInternal Medicine, 2001). Some trials fail to provide the reader with sufficient information to determine how the current results relate to other research. For example, Drew and Davies (2001) report that "Ginkgo biloba extract LI 1370 had no greater therapeutic effect than placebo in treating tinnitus. In addition, other symptoms of cerebral insufficiency were not significantly affected by the treatment (Table 3). The results from this trial are similar to some reports and contrast with others. 2 This study differs from other trials in many ways." (p.5). This provides the reader with little information from which to judge the efficacy of the product currently tested relative to other products that have been tested. Different botanical products can have different constituents and therefore differing therapeutic effects (Harkey et al., 2002; Gurley et al., 2000; Liberti & Der Marderosian, 1978; Groenewegen & Heptinstall 1986; Heptinstall et al., 1992; Nelson et al., 2002; Zhang et al., 2002). It is suggested here that discussion sections of trials of botanical interventions include a discussion of the trial results in the context of previous research while offering explicit consideration of the similarities or differences between products used therein. It is inappropriate to support or refute a trial's results by referring to literature that has tested a different product. Authors should be careful to clearly report when they are drawing inferences between heterogeneous products. When clinical trials on the specific product tested do not exist, pre-clinical data should be discussed. This includes animal, in vitro and other data.
COMMENTS Randomized allocation is the best tool to control for bias and confounding in controlled trials testing clinical interventions. Investigators must be sure to include in reports of these trials information that is required by the
CONSORT for Herbal Medicine Trials
205
reader to judge the validity and implications of the findings. In part, complete reporting of trials will allow clinician's to accurately appraise studies so as to modifY their clinical practice to reflect current evidence. The CONSORT statement was developed to assist investigators, authors, reviewers and editors on the necessary information to be included in reports of controlled clinical trials. The CONSORT statement is applicable to any intervention, including herbal medicinal products. Controlled trials of herbal medicines interventions do not adequately report the information suggested in CONSORT. Recently, several CONSORT items were elaborated to become relevant and complete for controlled trials of herbal medicines (Gagnier et al., 2006b). We expect that these recommendations will lead to more complete and accurate reporting of herbal medicine trials. We wrote this explanatory document to further explain the suggested elaborations and to assist authors in using them. We provide the CONSORT items and the associated elaborations, together with examples of good reporting and empirical evidence, where available. These recommendations for the reporting of RCTs of herbal medicine are open to change and revision as more evidence accumulates and critical comments are collected.
ACKNOWLEDGEMENTS The contributing papers included several additional authors that were instrumental in the development of concepts included in this chapter: Heather Boon, David Moher, Joanne Barnes, Paula Rochon and Claire Bombardier. We would like to thank Greer PaHoo for aiding in the preparation for the June meeting and Jaime DeMelo and Cyndi Gilbert for assisting Joel Gagnier and Claire Bombardier during the meeting procedures.
REFERENCES Altman, D.G., Schulz, KF. and Moher, D. et al. (2001). The revised consort statement for reporting randomized trials: Explanation and elaboration. Ann Internal Med., 134(8): 663-94. American Herbal Pharmacopoeia. http://www.herbal-ahp.org!order_online.htm Accessed Nov lBt, 2004. Annals of Internal Medicine. Information for authors. Available at: http://www.annals.org!. Accessed 10 January 200l. Association of Analytical Communities (AOAC). http://www.aoac.org! Accessed Nov 1st, 2004. Bailey, N.J., Sampson, J., Hyands, P.J., Nicholson, J.K and Holmes, E. (2002). Multicomponent metabolic classification of commercial feverfew preparations via high-field IH-NMR spectroscopy and chemometrics. Planta Med., 68(8): 734-8. Bauer, R. and Tittel, G. (1996). Quality assessment of herbal preparations as a precondition of pharmacological and clinical studies. Phytomedicine, 2(3): 193-8. Begg, C.B., Cho, M.K, Eastwood, S., Horton, R., Moher, D., OIkin, I., Pitkin, R., Rennie, D., Schulz, KF., Simel, D. and Stroup, D.F. (1996), Improving the quality of reporting of randomized controlled trials: The CONSORT statement. JAMA, 76(8): 637-39.
206
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Bent, S., Xu, L., Lui, L., Nevitt, M., Schneider, E., Tian, G., Guo, S. and Cummings, S. (2003). A randomized controlled trial of a Chinese herbal remedy to increase energy, memory, sexual function and quality of life in elderly adults in Beijing, China. Am J. Med., 115: 441-7. Betz, J.M., Eppley, R.M., Taylor, W.C. and Andrzejewski, D. (1994). Determination of pyrrolizidine alkaloids in commercial comfrey products (Symphytum sp.). J. Pharm Sci., 83(5): 649-53. Brinkhaus, B., Hummelsberger, J., Kohnen, R., Seufert, J., Hempen, C.H., Leonhardy, H., Nogel, R., Joos, S., Hahn, E. and Schuppan, D. (2004). Acupuncture and Chinese herbal medicine in the treatment of patients with seasonal allergic rhinitis: a randomized-controlled clinical trial. Allergy, 59(9): 953-60. Campbell, M.K., Elbourne, D.R. and Altman, D.G. (2004). for the CONSORT Group. CONSORT statement: extension to cluster randomized trials. BMJ, 328: 702-8. Carlson, M. and Thompson, R.D. (1998). Liquid chromatographic determination of methylzanthines and catechins in herbal preparations containing guarana. J AOAC Int., 81(4): 691-70l. Chan, K (2003). Some aspects of toxic contaminants in herbal medicines. Chemosphere, 52(9): 1361-71. Dowling, E.A., Redondo, D.R., Branch, J.D., Jones, S., McNabb, G. and Williams, M.H. (1996). Effect of eleutherococcis sneticosus on submaximal and maximal exercise performance. Med. Sci. in Sports Exercise, 28(4): 482-9. Dreikorn, K, Borkowsi, A., Braeckman, J. et al. (1998). Other medical therapies. In: Denis, L., Griths, K, Khoury, S., Cockett, A.T.K, McConnell, J., Chatelain, C., Murphy, G. and Yoshida, O. (eds) Proceedings of the 4th international consultation on benign prostatic hyperplasia (BPH). Plymbridge Distributors, Plymouth. 633-59. Dreikorn, K, Lowe, F., Borkowski, A., Buck, C., Braeckman, J., Chopin, D., Denis, L., Ferrari, P., Habib, F.K and Perrin, P. (2001). Other medical therapies. In: Chatelain, C., Denis, L., Foo, K.T., Khoury, S., McConnell, J. (eds) Proceedings of the 5th international consultation on benign prostatic hyperplasia (BPH). Plymbridge Distributors, Plymouth, pp. 479-511 Prostate. 26: 133-9. Drew, S. and Davies, E. (2001). Effectiveness of Ginkgo biloba intreating tinnitus: double blind, placebo controlled trial. BMJ. 322: 1-6. Fletcher, R.H., Fletcher, S.W. and Wagner, E.H. (1996). Clinical Epidemiology: The essentials. Lippincott: Philidelphia. Gagnier, J.J. (2003b). Tanacetum parthenium for migraine prophylaxis: Evidence based guidelines. Unpublished Manuscript. Gagnier, J.J., Boon, H., Rochon, P., Barnes, J., Moher, D., Bombardier, C.B. for the CONSORT Group (2006). Reporting randomized controlled trials of herbal interventions: an elaborated CONSORT statement. Ann. Int. Med, 144: 364-7. Gagnier, J.J., DeMelo, J., Boon, H., Rochon, P. and Bombardier, C. (2006a). Reporting quality of controlled intervention trials of herbal medicine. Amer. J. Med., 119(9). Gagnier, J.J., VanTulder, M.W., Berman, B. and Bombardier, C. (2006). Herbal medicine for low back pain. Cochrane Library, Issue l. Gardner, C.D., ChatteIjee, L.M. and Carlson, J.J. (2001). The effect of a garlic preparation on plasma lipid levels in moderately hypercholesterolemic adults. Atherosclerosis, 154: 213-20. Garrara, J., Hamrs, S., Eberly, L.E. and Matiak, A. (2003). Variations in product choices of frequently purchased herbs: caveat emptor. Arch. Int. Med., 163(19): 2290-5. German Federal Institute for Drugs and Medical Device (1999). The complete German Commission E monographs: Therapeutic guide to herbal medicines. Integrative Medicine Communications: MA. Greuter, W., Mcneill, J., Barrie, F.R., Burdet, H.M., Demoulin, V. and Filgueiras, T.S. et al. (1999). International Code of Botanical Nomenclature (ST LOUIS CODE), adopted by the Sixteenth International Botanical Congress, St Louis, Missouri, July. http:// www.bgbm.fu-berlin.de/iaptinomenclature/code/SaintLouis/OOOOSt.Luistitle.htm
CONSORT for Herbal Medicine Trials
207
Groenewegen, W.A. and Heptinstall, S. (1986). Amounts of feverfew in commercial preparations of the herb. Lancet, 1: 44-5. Gurley, B.J., Gardner, S.F. and Hubbard, M.A (2000). Content versus label claims in ephedracontaining dietary supplements. Am. J. Health-Syst Pharm., 57: 963-9. Haller, C.A, Duan, M., Benowitz, N.L. and Jacob, P. (2004). Concentrations of Ephedra alkaloids and caffeine in commercial dietary supplements. J. Anal. Taxicol., 28(3): 145-5I. Harkey, M.R, Henderson, G.L., Gershwin, M.E., Stern, J.S. and Hackman, RM. (2002). Variability in commercial Ginseng products: an analysis of 25 preparations. Am J. Clm. Nutr., 75(3): 600-I. Heptinstall, S., Awang, D.V. and Dawson, B.A et al. (1992). Parthenolide content and bioactivity of feverfew (Tanacetum parthenium [L.] Schultz-Bip.). Estimation of commercial and authenticated feverfew products. J Pharm Pharmacal, 44: 391-5. Ho, F.M., Hunag, P.J., Lee, F.K., Chern, T.S., Chiu, T.W. and Liau, C.S. (1999). Effect of acupuncture at Nei-Kuan on left ventricular function in patients with coronary artery disease. Am. J. of Chin. Med., 27(2): 149-56. Instructions to Authors. Phytomedicine: International Journal of Phytotherapy and Phytopharmacology. http://urbanfischer.de/journals/phytomed/instruc/Phytomi.htm Ioannidis,JohnP.A.,MD,Evans,StephenJ.w.,MSc,Gf<}tzsche,peterC.,MD, DrMedSci, O'Neill, Robert T., PhD, Altman, Douglas G., DSc, Schulz, Kenneth, PhD, Moher, David, PhD, for the CONSORT Group. (2004). Improving the reporting of harms in randomized trials: Expansion of the CONSORT statement. Ann. Int. Med., 141: 781-8. Isaacsohn, J.L., Moser, M., Stein, E.A, Dudley, K., Davey, K., Liskov, E. and Black, H.R (1998). Garlic powder and plasma lipids and lipoproteins: A multicenter, randomized, placebo-controlled trial. Arch. Int. Med., 158: 1189-1194. Jadad, AR, Moore, A and Carroll, D. et al. (1996). Assessing the quality of reports of randomized clinical trials: Is blinding necessary? Control Clin Trials, 17: 1-12. Johnson, E.S. (1984). Feverfew: a traditional remedy for migraine and arthritis. London: Sheldon Press. Johnson, E.S., Kadam, N.P., Hylands, D.M. and Hylands, P.J. (1985). Efficacy offeverfew as prophylactic treatment of migraine. BMJ, 291: 569-573. Kronenberg, F., Molholt, P., Zeng, M.L. and Eskinazi, D. (2001). A comprehensive information resource on traditional, complementary, and alternative medicine: toward an international collaboration. J. Alt. Complement. Med., 7: 723-729. Lefebvre, T., Foster, B.C., Drouin, C.E., Krantis, A, Livesy, J.F. and Jordan, S.A (2004). In VItro activity of commercial valerian root extracts against human cytochrome P450 3A4. J. Pharm Pharm Sci., 7(2):265-73. Liberti, L.E. and Der Marderosian, A. (1978). Evaluation of commercial ginseng products. J. Pharm Sci., 67: 1487-9. Linde, K., Jonas, W.B., Melchart, D. and Willich, S. (2001a). The methodological quality of randomized controlled trials of homeopathy, herbal medicines and acupuncture. Int. J. of Epidem, 30: 526-3I. Linde, K., ter Riet, G., Hondras, M., Vickers, A., Saller, Rand Melchart, D. (2001b). Systematic reviews of complementary therapies - an annotated bibliography. Part 2: Herbal medicine. BMC Complement Altern Med., 1(1): 5. Little, C. and Parsons, T. (2000). Herbal therapy for treating rheumatoid arthritis. Cochrane Library, 4. MacPherson, H., White, A, Cummings, M., Jobst, K., Rose, K. and Niemtzow, R (2001). Standards for Reporting Interventions in Controlled Trials of Acupuncture: The STRICTA Recommendations. Acupuncture in Medicine, 20(1): 22-5. Manning, J. and Roberts, J.C. (2003). Analysis of catechin content of commercial green tea products. J. Herb Pharacather., 3(3): 19-32. McGregor, B. (2002). Medical indexing outside the National Library of Medicine. J. Me Libr. Assoc., 90: 339-4I. Mix, J.A and Crews, W.D. (2002). A double-blind, placebo-controlled, randomized trial of Ginkgo biloba extract EGb 7611 in a sample of cognitively intact older adults: neuropsychological findings. Hum Psychopharmacol Clm. Exp., 17: 267-277.
208
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Moher, D., Jadad, A.R. and Tugwell, P. (1996). Assessing the quality of randomized controlled trials: Current issues and future directions. Int. J. Technol. Assessment Health Care, 12: 195-208. Moher, D., Sampson, M., Campbell, K, Beckner, W., Lepage, L., Gaboury, 1. and Berman, B. (2002). Assessing the quality of reports of randomized trials in pediatric and complementary and alternative medicine. BMC Pediatrics, 2. Moher, D., Schulz, KF. and Altman, D. for the CONSORT Group (Consolidated Standards of Reporting Trials). (2001). The CONSORT statement: revised recommendations for improving the quality of reports of parallel-group randomized trials. JAMA. 285: 198791. Morgenstern, C. and Biermann, E. (2003). The efficacy of Ginkgo special extract Egb 761 in patients with tinnitus. Int. J. Clin. Pharmacol. Ther., 5: 188-97. Mulrow, C.D., Cook, D.J. and Davidoff, F. (1998). Systematic Reviews: Critical Links in the Great chain of evidence. In; Systematic Reviews: Synthesis of Best Evidence for Health-care decisions, Mulrow, C. & Cook, D. (Eds), ACP: Phil. Murphy, J.J., Heptinstall, S. and Mitchell, J.RA. (1988). Randomized double-blind placebocontrolled trial of feverfew in migraine prevention. Lancet, 2(8604): 189-192. Murphy, LB., Reinsch, S., Najm, W.I., Dickerson, V.M., Seffinger, M.A., Adams, A. and Mishra, S. (2003). Searching biomedical databases on complementary medicine: the use of controlled vocabulary among authors, indexers and investigators. BMC Compl. Alt. Med., 3: 3. Nelson, M.H., Cobb, S.E. and Shelton, J. (2002). Variations in parthenolide content and daily dose of feverfew products. Am. J. Health-Syst Pharm., 59(16): 1527-31. Oberlies, N.H., Kim, N.C., Brine, D.R, Collins, B.J., Handy, RW., Sparacino, C.M. and Wani, M.C. et al. (2004). Analysis of herbal teas made from the leaves of comfrey (Symphytum officinale): reduction of N-Oxides results in order of magnitude increases in the measurable concentration of pyrrolizidine alkaloids. Public Health Nutr. 7(7): 919-24. Palevitch, D., Earon, G. and Carasso, R (1997). Feverfew (Tanacetum parthenium) as a prophylactic treatment for migraine: a double-blind placebo-controlled study. Phytotherapy Research, 11: 508-11. Patora, J., Majda, T., Gora, J. and Klimek, B. (2003). Variability in the content and composition of essential oil from lemon balm (Melissa offclnalis L.) cultivated in Poland. Acta Pol Pharm., 60(5): 395-400. Pellati, F., Benvenuti, S., Margro, L., Melegari, M. and Soragni, F. (2004). Analysis of phenolic compounds and radical scavenging activity of Echinacea spp. J. Pharm Biomed Anal., 35(2): 289-301. Pietri, S., Seguin, J.R, d'Arbigny, P., Drieu, K. and Culcasi, M. (1997). Ginkgo biloba extract (EGb 761) pretreatment limits free radical-induces oxidative stress in patients undergoing coronary bypass surgery. Cardiovasc Drugs Ther, 11: 121-31. Pietri, S., Seguin, J.R, d'Arbigny, P., Drieu, K. and Culcasi, M. (1997). Ginkgo bi/oba Extract (EGb 761) Pretreatment limits free radical-induced oxidative stress in patients undergoing coronary bypass surgery. Cardiovasc Drugs Ther, 11: 121-31. Piscitelli, S.C. and Burstein, A.H. (2002). Herb-drug interactions and confounding in clinical trials. J. Herb Pharacother, 2(1): 23-6. Roberts, C. (1999). The implications of variation in outcome between health professionals for the design and analysis of randomized controlled trials. Stat Med., 18: 2605-15. Sackett, D.L., Richardsom, W.S. and Rosenberg, W. et al. (1998). Evidence-Based Medicine: How to practice and teach EBM. Churchill Livingston: NY. Savulescu, J., Chalmer, 1. and Blunt, J. (1996). Are research ethics committees behaving unethically? Some suggestions for improving performance and accountability. BMJ. 313: 1390-3. Schulte-Lobbert, S., Holloubek, G., Muller, W.E., Schubert-Zsilavecz, M. Wurglics, M. (2004). Comparison of the synapotosomal uptake inhibition of serotonin by St. John's wort products. J. Pharm Pharmacol., 56(6): 813-8. Schulz, KF., Chalmers, 1., Hayes, RJ. and Altman, D.G. (1995). Empirical evidence of bias: Dimensions of methodological quality associated with estimates of treatment effects in controlled trials. JAMA, 273: 408-12.
CONSORT for Herbal Medicine Trials
209
Shelton, R.C., Keller, M.B., Gellenberg, A., Dunner, D.L., Hirschfeld, R. and Thase, M.E. et al. (2001). Effectiveness of St. John's wort in Major Depression: A randomized controlled trial. JAMA, 285: 1978-86. Sievenpiper, J.L., Arnason, J.T., Leiter, L.A. and Vuksan, V. (2003). Variable effects of American ginseng: a batch of American ginseng (Panax quinquefolius L.) with a depressed ginsenoside profile does not affects postprandial glycemia. Eur. J. Clin Nutr., 57: 243-8. Swanson, C.A. (2002). Suggested guidelines for articles about botanical dietary supplements. Am. J. Clin. Nutr, 75: 8-10. The CONSORT Group. http://www.consort-statement.orglendorsements/endorsements.htm Accessed Nov 1st: 2004. United States Pharmacopoeia. http://www .uspverified.orglresourcerequestform_man.html Accessed Nov 3rd, 2004. Vickers, A.J. and de Craen, A.J.M. (2000). Why use placebos in clinical trials? A narrative review of the methodological literature. J. Clin Epi., 53: 157-6l. Vuksan, V., Sivenpiper, J.L., Wong, J., Xu, Z., Beljan-Zdravkovic, U. and Arnason, J.T. et al. (2001). American ginseng (Panax quinquefohus L.) attenuates postprandial glycemia in a time-dependent but n dose dependent manner in healthy individuals. Am. J. Clin. Nutr., 73: 753-8. Vuksan, V., Starva, M.P., Sivenpiper, J.L., Koo, V.Y.Y., Wong, E. and Beljan-Zdravkovic, U. et al. (2000). American Ginseng improves glycemia in Individuals with normal glucose tolerance: Effect of Dose and Time escalation. J Amer Coll Nurtr, 19(6): 738-44. Weber, H.A., Zart, M.K., Hodges, A.E., Molloy, H.M., O'Brien, B.M. and Moody, L.A. et al. (2003). Chemical comparison of goldenseal (Hydrastis Canadensis L.) root powder from three commercial suppliers. J. Agric. Food Chem., 51(25): 7352-8. Xueping, Z., Zhongying, Z., Miaowen, J., Hong, W., Mianhua, W., Yaohong, S. and Haibo, C. (2004). Clinical study of Qingluo Tongbi Granules in treating 63 patients with rheumatoid arthritis ofthe type yin-deficiency and heat in the collaterals. J. Trad. Chin. Med., 24(2): 83-7. Zhang, H., Yu, C., Jia, J.y', Leung, S.W., Siow, Y.L., Man, R.Y. and Zhu, D.Y. (2002). Contents of four active components in different commercial crude drugs and preparations of danshen (Salvia miltiorrhiza). Acta Pharmacol Sin., 23(12): 1163-8.
"This page is Intentionally Left Blank"
8 Anxiolytic Activity Screening Studies on Extracts of a Few Medicinal Plants
ABSTRACT Aethusa cynapium, Artemisia abrotanum, Cimicifuga racemosa and Clematis erecta have been used traditionally in anxiety and sleep disorders but these plants have not been investigated so far to substantiate these therapeutic claims. The present study was aimed at screening the anxiolytic potential of the four plants. Petroleum ether, chloroform, methanol and water extracts of the plants have been evaluated for their anxiolytic activity using the elevated plus-maze model in mice. The methanol extract of Aethusa cynapium has demonstrated significant anxiolytic activity as compared to the standard drug diazepam. These results indicate that it is worth undertaking in depth investigation on Aethusa cynapium with a view to develop a potentially useful anxiolytic drug. Key words : Aethusa cynapium, Artemisia abrotanum, Cimicifuga racemosa, Clematis erecta, anxiolytic activity
INTRODUCTION Anxiety is a normal emotional response to real or potential danger. If anxiety becomes excessive, out of proportion and interferes with everyday life, management becomes necessary. The conventional treatment of anxiety involves use of medication like benzodiazepines, selective serotonin reuptake inhibitors (SSRls) etc or psychological therapy or a combination 1. Department of Pharmaceutical Sciences and Drug Research, Punjabi University,
Patiala, India. 2. #Pharmacognosy Division, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India (# - Address for correspondence). * Corresponding author : E-mail:
[email protected]
212
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
of the two. Common limitations of antianxiety drug therapy include comorbid psychiatric disorders and increase in dose leading to intolerable side effects (Cates et al., 1996). This has led researchers worldwide to investigate plants which are commonly employed in traditional and alternative systems of medicine for sleep disorders and related diseases (Farnsworth, 1980). Various plants have been investigated for their anxiolytic effects (Carlini, 2003). Valeriana species such as V. walliichii, V. officinalis, V. eduli, V. thalictroides have demonstrated antianxiety effect in human beings (Handa, 1992), Piper methysticum is employed clinically for the treatment of anxiety (Pepping, 1999; Reeder & Cupp, 2000; Billia et al., 2002). Numerous plants like Nardostachys jatamansi (Handa, 1992), Passiflora species like P. incarnata (Dhawan et al., 2001, 2001a), P.alata (Petry et al., 2001), Citrus aurantium (Carvalho-Frietas et al., 2002), Scutellaria lateriflora (Awad et al., 2003), Stachys lavandulifolia (Rabbani et al., 2003), Ginkgo biloba (Kuribara et al., 2003), Coriandrum sativum (Emamghoreishi et al., 2005), Turnera aphrodisiaca (Kumar & Sharma, 2005, 2005a), Centella asiatica (Wijeweera et al., 2006), Adiantum tetraphyllum (Bourbonnais-Spear et al., 2007), Apocynum venetum (Grundmann et al., 2007), have shown antianxiety activity in experimental animals. In the present study four plants, used in the traditional and complementary medicine have been investigated for their anxiolytic potential.
Aethusa cynapium Linn. (Umbelliferae) commonly called Fool's Parsley, has been used traditionally for gastrointestinal complaints in children, infantile cholera, summer diarrhoea, convulsions, sleep disorders and delirium (Flemming, 2000). In the homeopathic system of medicine, A. cynapium is used for milk intolerance in children, pylorus cramps and acute diarrhoea with vomiting (Vikramaditya & Joshi, 1997). A. cynapium contains alkaloid (Vikramaditya & Joshi, 1997), polyacetylenes (Andreev et al., 2001), essential oil, flavone glycosides and ascorbic acid (Flemming, 2000). Artemisia abrotanum Linn. (Compositae) commonly called Southernwood (Good, 1974), has been used traditionally for the treatment of digestive ailments, worms, menstrual disorders, marasmus, diarrhoea (www.herbs2000.com, 2000) and to relieve mental tension (www.herbs for life, 2003). A. abrotanum contains antimalarial sesquiterpene isofraxidin coumarin ethers (Greger et al., 1983; Cubukcu et al., 1990), essential oil (Mucciarelli et al., 1995), hydroxy coumarins (Elzbieta et al., 1984), spasmolytic flavonoid glycosides (Bergendorff & Sterner, 1995), peroxysemiketals (Ruecker et al., 1993) and choleretic phenolic acids (Swiatek et al., 1998).
Anxiolytic Activity Studies on Extracts of Plants
213
Cimicifuga racemosa Linn. (Ranunculaceae; synonym Actea racemosa) commonly known as Black Cohosh (Gleason & Cronquist, 1991), has been traditionally used to treat diarrhoea, sore throat, rheumatism, painful menstrual periods, menopausal disorders (Lieberman, 1998) dysmenorrhea, dyspepsia and snakebites (Bisset, 1989). The herb is available commercially in combination with St. John's Wort for depressive moods associated with premenstrual and menopausal symptoms (Fleming, 2000). The tincture of C. racemosa is used as anxiolytic, sedative emmenagogue and antitussive (Vikramaditya & Joshi, 1997). Remifemin® is a standardized extract ofthe plant available in Germany for management of menopause syndrome (Bisset, 1989). The plant is a hormone-free phytomedicine for treatment of climacteric symptoms related to menopause (Freudenstein et al., 2000; Liu et al., 2001; Kennelly, 2002). C. racemosa is rich in triterpene glycosides (Bedir & Khan, 2000; Wende et al., 2001; Chen et al., 2002, 2002a), 15,16-seco-cycloartane glycosides and tetra nor cycloartane glycosides (Nishida et al., 2003, 2003a), phenylpropanoid esters, aromatic acids, isoflavone and metals (Akihisa, 1999; Panossian et al., 2004). Clematis erecta Linn. (Ranunculaceae; synonym Clematis recta) is employed traditionally for disorders of skin, lymphatic system and genitourinary organs especially for gonorrhea. The tincture of C. erecta, an official preparation in the Pharmacopoeia Homeopathica Polyglottia 1872, is employed for the symptomatic relief of anxiety, nervous tension, insomnia, stress, fear, panic, colds, flu, fever and minor injury (Llyod & Lloyd, 1887; Felter & Llyod, 1898). C.erecta is reported to contain quaternary aporphine alkaloid (Slavik et al., 1987, 1995). MATERIALS AND METHODS
Plants Ariel parts ofAethusa cynapium, Artemisia abrotanum, Cimicifuga racemosa and Clematis erecta were collected from a cultivated source (Rati Ram Nursery) at village Khurrammpur via Kalsia, district Saharanpur (U.P., India) in March 2005. The identities of the plants were confirmed at the Museum-cum-Herbarium of the University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh.
Tested material Petroleum ether, chloroform, methanol and water successive Soxhlet extracts of the dried aeriel parts of the four plants viz. Aethusa cynapium, Artemisia abrotanum, Cimicifuga racemosa and Clematis erecta were prepared. Yields (as %, dry weight) are detailed in Table 1.
214
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Table 1. Percentage yields of extracts of the four investigated plants Plant extract Pet. Ether Chloroform Methanol Water
Aethusa cynapium 2.2 1.8 5.8 2.9
Yields (%, dried weight) Artemisia Clematis abrotanum erecta 1.3 1.3 9.1 3.9
0.5 3.6 12.1 10.3
Cimicifuga racemosa 2.5 2.8 12.8 15.6
Studied Activity Anxiolytic activity was evaluated using modified elevated plus-maze (Montgomery, 1958; Pellow et al., 1985; Kulkarni & Reddy, 1996). Vehicle [5% Tween 80 in Simple Syrup LP. (66.7% w/w sucrose in water)], test extracts and diazepam (reference drug), both suspended in vehicle, were administered orally 45 min prior to the antianxiety test, and the behavior of animals on the elevated plus-maze was recorded for 5 min in terms of (i) number of entries into open or closed arms (ii) average time spent by the mouse in open or closed arms.
Animals Swiss albino mice of either sex, weighing 20-24 g were procured from the Central Animal House of the Panjab University, Chandigarh. The mice were allowed standard laboratory feed and water ad libitum. Groups of five mice were used in all sets of experiments. Animals were fasted overnight before use. All animal studies were carried out as per the guidelines of the Institute Ethical Committee, Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala.
Statistical Analysis Analysis of variance (AN OVA) and the Tukey's multiple range tests was employed to determine statistical significance. Difference were considered significant at p<0.05. RESULTS AND CONCLUSIONS Aethusa cynapium, Artemisia abrotanum, Cimicifuga racemosa and Clematis erecta have been used traditionally in anxiety and sleep disorders but these plants have not been investigated so far to substantiate these therapeutic claims. The present study was carried out to validate the traditional claims. Different extracts of the four plants have been prepared using solvents in increasing order of polarity. This is done in order to extract completely non polar to completely polar constituents from the plant material. The anti anxiety activity of these extracts were compared with
Anxiolytic Activity Studies on Extracts of Plants
215
the activity of the standard drug diazepam and the control using the elevated plus-maze model. The elevated plus-maze, is a novel test for the selective identification of anxiolytic drug effects in rodents (Vogel, 2002). The elevated plus-maze model is effective, cheap and simple, requires no preliminary training for the mice, and does not cause much discomfort to them while handling. The fear due to height (acrophobia) induces anxiety in mice when placed on the elevated plus-maze. The ultimate manifestation of anxiety and fear then is exhibited by decrease in motor activity, which is measured by the time spent by mice in the open arms. Anxiolytic compounds, by decreasing anxiety, increase the open arm exploration time, anxiogenic compounds have the opposite effect (Kulkarni, 2002). The mean time spent by the mice in open arms of the EPM after oral administration of various doses of the different extracts of the four plants is reported in Table 2. Table 2. Anxiolytic effects of extracts of the four investigated plants using the Elevated Plus Maze model Comparison with Average Time spent in open arms (seconds) a Control (Vehicle, 0.25 mL) 3.2 ± 1.3 Standard (Diazepam, 2 mg/kg) 12.2 ± 1.2 Treatment
Average Time spent in open arms (seconds)a Dose (mg!kg), Aethusa Artemisia Cimicifuga Clematis p.o cynapium abrotanum erecta racemosa
Pet.Ether Extract
50 100 200 400
5.7 6.8 7.9 5.9
± ± ± ±
1.0*'** 0.4*·** 0.8*·** 0.9*·**
4.5 3.2 7.9 4.9
± ± ± ±
0.5 *.** 0.4* 0.9*·** 0.5*·**
1.7 1.1 1.6 3.2
± ± ± ±
0.1*·** 0.1*·** 0.2*·** 0.4*
2.3 2.7 3.3 2.6
± ± ± ±
0.2* 0.2* 0.4* 0.2*
Chloroform extract
50 100 200 400
5.1 8.1 7.6 7.0
± ± ± ±
1.2*·** 1.2*'** 1.0*·** 1.3*·**
2.9 4.3 2.6 2
± ± ± ±
0.2* 0.5* 0.2* 0.2*·**
1.0 1.6 1.4 2.1
± ± ± ±
0.1*·** 0.2*·** 0.2*·** 0.2*·**
1.7 2.6 0.8 3.3
± ± ± ±
0.1*·** 0.2* 0.2*·** 0.4*
Methanol extract
50 100 200 400
7.7 8.2 9.5 12.7
± ± ± ±
0.9*·** 0.9*'** 1.1*·** 1.5**
0.9 1.6 1.1 6.9
± ± ± ±
0.1*·** 0.1*·** 0.1*·** 0.7*·**
0.6 1.0 1.9 5.6
± ± ± ±
0.1* 0.1* 0.1* 0.6*
2.6 2.5 4.7 1.2
50 100 200 400
5.2 8.2 8.4 7.9
± ± ± ±
0.6*'** 1.7*·** 1.5*·** 1.4*·**
0.8 0.7 0 8.3
± ± ± ±
0.1*·** 0.1*·** 0.0*·** 1.0*·**
2.2 ± 1.5 ± 2.7 ± 1.15±
0.3* 1.8*·** 0.3* 0.2*·**
0.5 0.9 1.2 0.7
Water extract
± 0.2* ± 0.2*
± 0.5*'** ± 0.1*·** ± ± ± ±
0.1*'** 0.1*·** 0.1*·** 0.1*·**
a:Values are expressed as mean ± S.D. (n = 5). ANOVA followed by Tukey's multiple range test. p<0.05. * =significant with respect to standard, ** = significant with respect to control.
216
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Animals treated with diazepam (2 mg/kg, p.o.) spent 12.2 sec in open arms of the plus maze and the control group animals spent 3.2 sec. Of the four extracts of Aethusa cynapium, the methanol extract shows a dose dependent increase in motor activity, with the animals spending 12.7 seconds at 400 mg/kg in the open arms of the maze. This activity is statistically comparable to that of diazepam. The Artemisia abrotanum extracts, although significant at some doses when compared to the control, were not significant in comparison with diazepam. The four extracts of Clematis erecta and Cimicifuga racemosa do not show anxiolytic activity at any of the doses administered. The results of the present study show that of the four plants tested only Aethusa cynapium demonstrated significant anxiolytic activity, and indicate that it is worth undertaking in depth investigation on Aethusa cynapium with a view to develop a potentially useful anxiolytic drug.
REFERENCES Akihisa, T., Hideshima, R., Koike, K., Kimura, Y. and Nikaido, T. (1999). Cycloarta-16,24dien-3-ol: revised structure ofcimifugenol, a cycloartane triterpenoid. Chem. Pharm. Bull., 47: 1157- 1160. Andreev, G.N., Schulz, H., Schrader, B., Fuchs, R., Popov, S. and Handjieva, N. (2001). Nondestructive NIR-Ft-Raman analyses in practice. Partl. Analyses of plants and historic textiles. Fresenius J. Anal. Chem., 371(7): 1009-17. Awad, R., Amason, J.T., Trudeau, V., Bergeron, C., Budzinski, J.W., Foster, B.C. and Merali, Z. (2003). Phytochemical and biological analysis of skullcap (Scutellaria latenflora L.): a medicinal plant with anxiolytic properties. Phytomedicine, 10(8): 640-9. Bedir, E. and Khan, l.A.(2000) Cimiracemoside A: A new cyclolanostanol xyloside from the rhizome of Cimic~fuga racemosa. Chem. Pharm. Bull., 48: 425-427. Bergendorff, O. and Sterner, O. (1995). Spasmolytic flavonols from Artemisia abrotanum. Planta Medica, 61(4): 370-71. Billia, A.R., Gallori, S. and Vincieri, F.F. (2002). Kava-kava and anxiety:growing knowledge about the efficacy and safety. Life Sci., 70: 2581-97. Bisset, N.G. (1989). Herbal drugs and phytopharmaceuticals- A handbook for practice on a scientific basis, CRC press, 90-91. Bourbonnais-Spear N., Awad R., Merali, Z., Maquinc, P., Cal, V. and Amason, J.T. (2007). Ethnopharmacological investigation of plants used to treat susto, a folk illness. J. Ethnopharmacol., 109: 380-387. Carlini, E.A. (2003). Plants and the central nervous system. Pharmacol., Biochem. and Behavior, 75: 501-512. Carvalho-Freitas, M.l. and Costa, M. (2002). Anxiolytic and sedative effects of extracts and essential oil from Citrus aurantium L. Biol Pharm Bull., 25(12): 1629-33. Cates, M., Wells, B.G. and Thatcher, G.W. (1996). Anxiety Disorders. In: Textbook of Therapeutics: Drug and Disease Management, HerFindal, E.T. and Gourley, D.R. (editors), 6 th edition, Williams and Wilkins, Maryland, USA, pp. 1073-93. Chen, S.N., Li, W., Fabricant, D.S., Lu, Z.Z., Fong, H.H.S. and Farnsworth, N.R. (2002) Cimiracemates A-D, phenylpropanoid esters from the rhizomes of Cimicifuga racemosa. J. Nat. Prod., 61: 409-413. Chen, S.N., Li, W., Fabricant, D.S., Lu, Z.Z., Fong, H.H.S. and Farnsworth, N.R. (2002a). Cimiracemosides I-P, new 9,19-cyclolanostane triterpene glycosides from Cimicifuga racemosa. J. Nat. Prod., 65: 1391-1397. Cubukcu, B., Bray, D.H., Warhurst, D.C., Mericli, A.H., Ozhatay, N., Sariyar, G. (1990). In v~tro antimalarial activity of crude extracts and compounds from Artemisia
Anxiolytic Activity Studies on Extracts of Plants
217
abrotanum L. Phytotherapy Research, 4(5): 203-4. Dhawan, K, Kumar, S. and Sharma, A. (2001). Anti-anxiety studies on extracts of Passiflora incarnata L. J. Ethnopharmacol., 78: 165-170. Dhawan, K., Kumar, S. and Sharma, A. (2001a). Anxiolytic activity of aerial and underground parts of Passiflora incarnata. Fitoterapia, 72(8): 922-6. Elzbieta, S., Barbara, 0., Piotr, G. and Krystyna, S. (1984). Pharmacologically active complex of hydroxyl coumarins from Christmas tree worm wood (Artemisia abrotanum L.). Pol. Pl., 120: 547. Emamghoreishi, M., Khasaki, M. and Aazam, M.F. (2005). Coriandrum sativum: evaluation of its anxiolytic effect in the elevated plus-maze. J. Ethnopharmacol. 96: 365-370. Farnsworth, N.R. (1980). The development of pharmacological and chemical research for application to traditional medicine in developing countries. J. Ethnopharmacol., 2: 173-18l. Felter, H.W. and Lloyd, J.U. (1898). King's American Dispensatory. Through http:// www.ibiblio.orglherbmed. Fleming, T. (2000). PDR for herbal medicmes, 2 nd edition, published by Medical economics company inc, Montvale, NJ07647-1742, pp. 316. Freudenstein, J., Dasenbrock, C. snd Nisslein, T. (2000) Lack of promotion of estrogen dependent mammary gland tumors in vivo by an isopropanolic black cohosh extract. Phytomed~cine, 7: 13. Gleason, H.A. and Cronquist, A. (1991). Manual of vascular plants of the North-eastern United States and adjacent Canada. 2nd edition, Bronx, New York Botanical Garden. Good, R. (1974). The geography of flowering plants. 4th edition, Longman, London. Greger, H., Hofer, 0., Nikiforov, A. (1983). New sesquiterpene- coumarin ethers from Achillea and Artemisia species. J. Natural Products, 45(4): 455-6l. Grundmanna, 0., Nakajima, Jun-Ichiro, Seo, S. and Butterweck, V. (2007). Anti-anxiety effects of Apocynum venetum L. in the elevated plus maze test. J. Ethnopharmacol., 110: 406-41l. Handa, S.S. (1992). Medicinal plants based drug industry and emerging plant drugs. Curro Res. Med. Aromat. Plants, 14: 233-262. http//www.herbs for life (2003). http//www.herbs2000.com (2000). Kennelly, E.J., Baggett, S., Nuntanakorn, P., Ososki, A.L., Mori, S.A., Duke, J., Coleton, M. and Kronenberg, F. (2002). Analysis of thirteen populations of black cohosh for formononetin. Phytomedicine, 9(5): 461-467. Kulkarni, S.K and Reddy, D.S. (1996). Animal behavioral models for testing antianxiety agents. Meth Find Exp Clm Pharmacol., 18: 219. Kulkarni, S.K (2002). Handbook of Experimental pharmacology (3 rd edition>. Vallabh Prakashan, Delhi, India., pp. 135-137. Kumar, S. and Sharma, A. (2005). Anti-anxiety activity studies of various extracts of Turnera aphrodisiaca Ward. J Herb Pharmacother., 5(4): 13-2l. Kumar, S. and Sharma, A. (2005a). Anti-anxiety Activity Studies on Homoeopathic formulations of Turnera aphrodisiaca. Evid. Based Complement. Alternat. Med., 2: 117-119. Kuribara, H., Weintraub, S.T., Yoshihama, T. and Maruyama, Y. (2003). An anxiolytic-like effect of Ginkgo biloba extract and its constituent, ginkgolide-A, in mice. J Nat Products, 66(10): 1333-7. Lieberman, S. (1998). A review of the effectiveness of Cimicifuga racemosa (black cohosh) J. Womens Health, 7:5: 525-9. Liu, Z., Yang, Z., Zhu, M. and Huo, J. (2001). Estrogenicity of black cohosh (C~micifuga racemosa) and its effect on estrogen receptor level in human cancer MCF-7 cells. Wei Sheng Yan Jiu ., 30: 77-80. Through L~fe Sciences, 2003:73: 1215-1229. Lloyd, J.U. and Lloyd, C.G. (1887). Drugs and medicines of North America. Through Henriette's Herbal Homepage. Montgomery, K.C. (1958). The relation between fear induced by novel stimulation and exploratory behaviour. J. Compo Physiol. Psychol., 48: 254-260.
218
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Mucciarelli, M., Caramiello, R. and Maffei, M. (1995). Essential oils from some Artemisia species growing spontaneously in north-west Italy. Flavour and Fragrance Journal, 10(1): 25-32. Nishida, M., Yoshimitsu, H., Nohara, T., Okawa, M. and Ikeda, T. (2003). Two new 15,16seco-cycloartane glycosides from Cimicifuga racemosa. Chem. Pharm. Bull., 51(10): 1215-1216. Nishida, M., Yoshimitsu, H. and Nohara, T. (2003a). Three cycloartane glycosides from Cimicifuga racemosa and their immunosuppressive activities in mouse allogenic mixed lymphocyte reaction. Chem. Pharm. Bull., 51(3): 354-356. Panossian, A., Danielyan, A., Mamikonyan, G. and Wikman, G. (2004). Methods of phytochemical standardization of rhizome Cimicifugae racemosae. Phytochem. Anal., 15(2): 100-8. Pellow, S., Chopin, Ph., File, S.E. and Briley, M. (1985). Validation of open:closed arm entries in an elevated plus maze as a measure of anxiety in the rat. J. Neurosci. Meth., 14: 149-167. Pepping, J. (1999). Kava: Piper methysticum. Am. J. Health- syst Pharm., 56: 957-63. Petry, R.D., Reginatto, F., de-Paris, F., Gosmann, G. and Salguciro, J.B. (2001). Comparative pharmacological study of hydroethanol extracts of Passiflora alata and P. edulis leaves. Phytother. Res., 15: 162-4. Rabbani, M., Sajjadi, S.E. and Zarei, H.R. (2003). Anxiolytic effects of Stachys lavandulifolia Vahl on the elevated plus-maze model of anxiety in mice. J Ethnopharmacol., 89(2-3): 271-6. Reeder, S. and Cupp, M.J. (2000). Kava. In: Toxicology and Clinical Pharmacology of Herbal Products. Cupp, M.J. (Editor), Humana Press, New Jersey. Ruecker, G., Manns, D. and Wilbert, S. (1993). Peroxides as plant constituents XII Davanone type peroxides from southernwood (Artemisw abrotanum) and their preparations. Arch. Pharm., 326(8): 457-60. Slavik, J., Bochorakova, J. and Slavikova, L. (1987). Occurrence of magnofloorine and corytuberine in some wild or cultivated plants of Czechoslovakia. Collect Czech Chem Commun., 52: 804-812. Slavik, J. and Slavikova, L. (1995), Quaternary isoquinoline alkaloids and some diterpenoid alkaloids in plants of the Czech Republic. Collect Czech Chem Commun., 60: 1034-1041. Swiatek, L., Grabiar, B. and Kalemba, D. (1998) Phenolic acids in certain medicinal plants of the genus Artemisia. Pharm. Pharmacol. Lett., 4: 158-60. Vikramaditya Joshi, P. (1997). A guide to important medicinal plants used in homeopathy, Homeopathic Pharmacopoeial laboratory, V. 2, pp. 23 and pp. 4. Vogel, H.G. (2002). Drug Discovery and Evaluation Pharmacological assays (Ed) second edition. Springer-Verlag, Berlin, Germany., pp. 434-435. Wende, K., Mugge, C., Thurow, K., Schopke, T. and Lindquest, U. (2001). Actaeaepoxide 3O-b-D- xylopyranoside, a new cycloartane glycoside from the rhizomes of Actaea racemosa (Cimicifuga racemosa). J. Nat. Prod., 64(7): 986-989. Wijeweeraa, P., Arnasona, J.T., Koszyckib, D. and Merali, Z. (2006). Evaluation of anxiolytic properties of Gotukola - (Centella asiatica) extracts and asiaticoside in rat behavioral models. Phytomedicine, 13: 668-676.
9 Study of Eugenia jambolana and Momordica charantia in Respect to Safety and Antidiabetic Effect In vivo and In vitro YELE, SANTOSH 1 AND VEERANJANEYULU, A.l,*
ABSTRACT Diabetes mellitus is composed of a myriad of derangements on carbohydrate, lipid and protein metabolism, which are associated to an absolute or relative deficiency of insulin secretion / action. Out of a large number of herbal drugs in the Ayurvedic system of medicine of India, Eugenia jambolana of family Myrtaceae and Momordica charantia from cucurbitaceae (Jamun) is being widely used to treat diabetes by the traditional practitioners over many years. They are prepared as an aqueous or ethanolic extract, by infusion or as a juice of the raw plant. MC has a long history of use as a folklore hypoglycaemic agent where the plant extract has been referred to as vegetable insulin Eugenia jambolina (EJ) is well known plant with identified antidiabetic, laxative, anti-inflammatory and antimicrobial properties in traditional literature. In the present study the plants have been explored for the following parameters like safety evaluation, antidiabetic properties and in vitro antidiabetic properties, individually as well as in combination (1: 1) to find out additive of synergistic effect. Acute oral toxicity of water extract of Eugenia jambolana was evaluated in albino mice of both sexes. These results show that the water extract of Eugenia jambolana is toxicologically safe by oral administration. The effects of Eugenia jambolana bark extracts were studied with alloxan induced diabetes model for antidiabetic property. Albino Wistar rats weighing 150-200 g (N = 6) were fed E. jambolana bark extract and combination of EJ + MC (50:50) 500 mgt kg of b.w. for 14 days. In another investigation, we have studied the effects 1. Department of Pharmacology, School of Pharmacy and Tech. Management NMIMS
University, Mumbai - 400 056 Maharashtra, India.
* Corresponding author: E-mail:
[email protected]
220
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
ofaqueous fruit extract ofMe and aqueous bark extract ofEJ on the transport of d-glucose, I-tyrosine across rat-everted intestine in vitro. Everted intestinal sacs from rats were mounted in an organ bath containing Krebs-Henseleit bicarbonate buffer. Graded concentrations (1.5-12 mg/mL) of Me and EJ extract were incubated in the mucosal solution. The serosal appearance and mucosal disappearance ofd-glucose, and I-tyrosine were significantly inhibited with increasing graded concentrations of Me and EJ extracts. The aqueous extract of the plants was found to inhibit primarily the uptake of glucose in a dose-dependent manner. Uptake of tyrosine was affected at high substrate concentrations only. It is hypothesized that bioactive phytochemicals such as sap on ins in Me fruit extract and tannins from EJ inhibits the active transport of d-glucose, I-tyrosine across rat intestine It is revealed that Me and EJ can be a potential alternative drug therapy of postprandial hyperglycemia via inhibition of glucose uptake across the small intestine and could involve a washout of glucose from the blood stream. Key words: Acute toxicity, intestinal uptake, Eugenia jamb alana, Momordica charantia
INTRODUCTION Diabetes mellitus is a group of metabolic disorders characterized by hyperglycemia. These metabolic disorders include alterations in the carbohydrate, fat and protein metabolisms associated with absolute or relative deficiencies in insulin secretion and/or insulin action. Insulin is the hormone produced in the p-cells of pancreas which enables the cells to absorb glucose from the blood and also help in the utilization ofthe glucose in the cells by glycolysis, tricarboxylic acid cycle, hexose monophosphate shunt, glycogenesis etc. In diabetes body fails to produce insulin and excess glucose accumulates in the blood instead of being utilized or stored. The characteristic symptoms of diabetes are polyuria, polydypsia, polyphagia, pruritus and unexpected weight loss etc. Diabetes can be classified into two types: (a) type 1 diabetes (insulin-dependent diabetes mellitus or IDDM) and (b) type 2 diabetes (non-insulin dependent diabetes mellitus or NIDDM) (Guyton, 2002). Exercise and having a controlled diet are recommended for the treatment of both types of diabetes. In addition, insulin is used to treat cases of type 1 diabetes. Oral hypoglycemic agents (OHA) such as sulfonylureas, biguanidines, thiazolidinediones and a-glucosidase inhibitors are often used to treat cases of type 2 diabetes. When therapy with oral hypo-glycemic agents is ineffective, insulin also can be used to treat type 2 diabetes (Liu & Wang, 1996; Zhao, 1999). Management of diabetes without any side effects is still a challenge to the medical system. There is an increasing demand by patients to use the natural products with antidiabetic activity, because insulin and ORAs are having so many side effects. Medicinal plants continue to provide valuable
E. jambolana and M. charantia in Respect to Safety and Antidiabetic Effect
221
therapeutic agents, both in modern medicine and in traditional system. The doubts about the efficacy and safety of the oral hypoglycemic agents have prompted a search for safer and more effective drugs in the treatment of diabetes (Reaven, 1983). A wide variety of the traditional herbal remedies are used by diabetic patients, especially in the third world countries (Lin, 1992; Ospinag & Serrano, 1995; Day, 1998; Gray & Flatt, 1998; Shimada et al., 1998) and may therefore, represent new avenues in the search for alternative hypoglycemic drugs. The folk medicine in India has described several kinds of ayurvedic herbs and plant crude drugs, belonging to various families to be concerned with the treatment of diabetes mellitus. Plant drugs are frequently considered to be less toxic and more free from side effects than synthetic ones (Pari & Umamaheswari, 2000). In the traditional system ofIndian medicine plant formulation and in several cases, combined extracts of plants are used as the drug of choice rather than individual. Presently several research laboratories are involved in isolating new herbal oral hypoglycemic agents or potent herbal formulation. The renounced OHA, Metformin which gained popularity in recent years got its shape from its ancestor, active ingredient Galegine or isoamylene guanidine from Galega officinalis (Cusi & DeFronzo, 1998). More than 100 medicinal plants are mentioned in the Indian system of medicines including folk medicines for the management of diabetes, which are effective either singly or in combinations. Many of these have shown promising effects (Kumari & Devi, 1993.). Various herbal formulations like D-400 (Mitra et al., 1996) and Trasina (Bhattacharya et al., 1997) are well known for their antidiabetic effects. While going through the previous works of different investigators, some variations in their experimental results have been observed. Incomplete extraction procedure, inadequate pharmacognosy and insensitive animal model may be the reasons for showing variable and even negative results. We have guarded against all these factors in our experiments and selected only two hypoglycaemic medicinal plants for our studies. Out of which one is Momordica charantia, traditionally and experimentally known for the antidiabetic activity (Grover et al., 2002) and another crude drug is Eugenia jambolana L. (Bark) less know part of plant and hardly explored for its use in diabetes as compared to its other parts like leaves, fruit and pulp (Nadakarni, 1954). Aim of present study is to confirm the ethnobotanical claim of antidiabetic property of Eugenia jambolana bark, and its use with momordica charantia. Folklore literature state that bark ofEJ and fruit powder ofMC exhibit the synergistic effect and control both blood glucose and body weight with short period of time as compared to individual drugs. Sahyadri hills row (Pune-Kolhapur region, Maharashtra, India), which lie geographically in the Western Ghats, are known for the rich heritage of the flora. A number of plants with known and unknown medicinal values
222
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
are available here, which have to be explored for their use in the effective treatment of diabetes mellitus as described in the folk literature. But no systematic scientific studies were carried out on these plants by that mean.
MATERIALS AND METHODS Plant Material The bark of Eugenia jambolana and fruits of Momordica charantia were collected from local farm of Baramati (Maharashtra) and Pune region of Western Ghats of Maharashtra, respectively. Both the plant parts were authenticated by the scientists of BSI (Botanical Survey of India) Pune, Maharashtra. After collection, Bark of EJ was dried under shade and then washed out debris and adhered matter and again dried to remove moistness of bark. Bark was applied to disintegrator to make the fine powder. Same way fruits of Me were shade dried after proper removal of seeds and washing with water and powdered to coarse size.
Preparation of Extracts For entire experimentation cold macerated extract of both the plants were used. Powdered bark of EJ and fruits of MC were subjected for cold maceration up to 48 h. At the end of maceration syrupy liquid of both the drugs were obtained, to make it free flow powder extract were concentrated and dried under the reduced pressure with the help of rotary vacuum evaporator (Rotavap®). The yield of crude extracts from EJ and MC was 16.6% and 33.1% respectively. Extract of EJ was used individually and in combination with MC in the proportion of (EJ+MC-50:50). Alloxan was purchased from SD fine chemicals Company Inc., while all other chemicals used in this experiment were purchased from SD fine chemicals.
Acute Toxicity of EJ Bark Swiss albino mice were used for oral acute toxicity study and divided into four groups, each containing five animals. The bark extract was administered orally at doses of 1000, 3000, and 5000 mg/kg body weight. Distilled water was administered to control group. The animals were evaluated at 0.5,1,2,4,24 and 48 h, after administration to assess possible clinical or toxicological symptoms. Signs of toxicity and mortality were observed daily for 13 days. At the end of 14th day after the last observation the mice were killed and the liver, lungs, heart, spleen and kidneys were withdrawn, weighed and carefully examined macroscopically for any abnormal, pathological signs of toxicity.
Experimental Animals for Antidiabetic activity Male wistar rats weighing 180-200 g were obtained from the Haftkins' Research Institute Mumbai. The rats were housed at a regulated
E. jambolana and M. charantia in Respect to Safety and Antidiabetic Effect
223
temperature (22°C) and humidity (55%) controlled room with a 12 h light: 12 h dark cycle. A water and standard pellet diet were available ad libitum throughout the experimental period.
Antidiabetic activity Effect of the crude extracts on serum glucose level in alloxan-induced diabetic rats Diabetes was induced by a single intravenous injection of 60 mg/kg of alloxan monohydrate (dissolved just before use in 0.9% NaCl) to overnight fasted rats (Silva et al., 2002 with modification). A serum glucose range of 400-550 mg/dl was used for the experiment. Animals in which the development of hyperglycemia was confirmed (around 90%), 72 h after the alloxan injection, were randomly allocated into four groups of five rats to each treatment: Group I, diabetic control received the saline solution Group II, non-diabetic received distilled water Group III, diabetic received the crude EJ extract 500 mg/kg b.w. Group IV, diabetic received the combined extracts EJ+MC (50:50) 500 mg/kg b.w Blood samples were collected from the retro-orbital plexus before the administration of extract or combination serum glucose levels were measured at zero time (before receiving the extract).
Effects of (MC+EJ) on transport of d-glucose and l-tyrosine across the intestinal sac - in vitro Adult male Swiss albino rats weighing 200-250 g maintained on commercial feed were used for this study. After overnight fasting, rats were killed by a severe blow on the head against a hard surface. The abdomen was opened by a midline incision. The whole of the small intestine was removed by cutting across the upper end of the duodenum and the lower end of the ileum and manually stripping the mesentery. The small intestine was washed out with normal saline solution (0.9% w/v NaCl) using a syringe equipped with blunt end. Intestinal segments (1062 cm) were then everted according to the method described by Wilson and Wiseman. After weighing, the empty sac was filled with 1 mL of Krebs- Henseleit bicarbonate buffer (KHB). The composition of the buffer was (mmll): NaHC0 3 25; NaCI 118; KCI 4.7; MgS0 4 1.2; NaH 2 P0 4 1.2; CaCl 2 1.2; and Na 2 EDTA 9.7 mg/l. Glucose (2 gil) was added to the medium just before the start of the appropriate experiments. The pH was maintained at 7.4. The sac was filled with a blunted-ended syringe and then slipped off the needle carefully and the loose ligature on the proximal end was tightened. After weighing, the
224
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
distended sac was placed inside an organ bath containing 50 mL of the same incubation medium. The organ bath was surrounded by a water jacket maintained at 37-40°C and placed in metabolic shaker at a frequency of 100-110 shakes/min. The incubation medium was continuously being bubbled with a mixture of95% 02 and 5% CO 2 , At the end of the incubation period (30 min.), the sacs were removed from the organ bath, blotted by a standardized procedure and weighted again. The serosal fluid was drained through a small incision into a test tube. The emptied sac was shaken gently to remove the adhered fluid and the tissue was weighted again. The weight of the empty sac before and after the incubation did not differ significantly. The initial serosal volume was determined as the difference between the weights ofthe empty and filled everted sac prior to incubation. The final serosal volume was calculated by subtracting (after incubation) the weight of the empty sac from that of the filled sac (Mahomoodally et al., 2005, 2006). The aqueous EJ extract individually and with MC extracts (50:50) were incubated in the mucosal solution in the organ bath. The active transport of d-glucose and I-tyrosine were evaluated by measuring the increase in concentration of both compounds inside and outside the intestinal sacs after 30 min. of incubation. L-Tyrosine in the incubating buffer solution was determined as described by Lowry et al. (1951) with modifications. Glucose was measured using a commercially available glucose oxidase kit. The terms used for d-glucose and I-tyrosine transfer are mucosal glucose transfer, serosal glucose transfer and gut glucose uptake. Mucosal glucose transfer is the amount of glucose that disappeared from the mucosal fluid while serosal glucose transfer is the amount of glucose that entered the serosal fluid. Gut glucose uptake is the difference in glucose concentration between the mucosal and serosal fluid after incubation.
RESULTS AND DISCUSSION Acute Oral Toxicity Acute oral toxicity study of Eugenia jambolana extract showed that no mortality of mice occurred, at the doses of 1000, 3000, and 5000 mg/kg body weight given orally. This is an indication that the extract has negligible level of toxicity when administered orally. Besides, no signs of toxicity, such as convulsion, vomiting, diarrhoea, paralysis, breathing difficulties, bleeding, restless, irritation, and abnormal posture, were observed in Eugenia jambolana extract-treated mice. Thus, it can be suggested that Eugenia jambolana extract is virtually nontoxic. In conclusion, Eugenia jambolana extract was found to be fairly nontoxic when oral acute toxicity study in mice was performed.
E. jambolana and M. charantia in Respect to Safety and Antidiabetic Effect
225
Effect of the crude extracts on serum glucose level in alloxan-induced diabetic rats Blood sugar level increased as expected in alloxan-injected animals, since alloxan causes a massive reduction in insulin release, by the destruction of the beta-cells of the islets of Langerhans and inducing hyperglycemia (Goldner et al., 1943). Oral administration of EJ (500 and mg/kg b.w.) and combined form of extract EJ+MC (500 mg/kg b.w.) resulted in a significant reduction in the blood glucose. In our study we have found that EJ decreases blood glucose in alloxan diabetic rats. In this context a number of other plants have also been observed to have hypoglycaemic effects. But when the results compared with individually action of EJ and its combination (EJ+MC) shows remarkably reduction in elevated blood glucose (Fig 1). The possible mechanism for hypoglycemic action may be potentiating the insulin effect of plasma by increasing either the pancreatic secretion of insulin from the ~-cells of islets of Langerhans or its release from bound insulin. As MC is suppose to known to posses insulin like compound and EJ posses tannins, triterpenes responsible for activity of reduction in plasma blood glucose. We have demonstrated that the folk medicinal plant EJ possesses a hypoglycemic effect. Further, active research is underway in our laboratory to elucidate the mechanisms of action of this medicinally important plant.
Effects of (Me + EJ) on transport of d-glucose and l-tyrosine across the intestinal sac - in vitro The effects of EJ crude aqueous extract individually and in combination with MC on d-glucose, I-tyrosine and transport (serosal appearance) across everted intestinal sacs of rat are summarized in (Tables 1 & 2). In order to find the lowest inhibitory concentration and any dose dependent relationship, graded concentrations of EJ and EJ+MC (from 1.5 to 12 mg/mL) were incubated with the intestinal segments in the mucosal solution. Data from the present investigation indicate that EJ inhibits significantly (p<0.05) the active transport of d-glucose, I-tyrosine. It was found that increasing graded concentrations of EJ+MC from 1.5 to 12.0 mg/mL in the mucosal solution decrease significantly the absorption as well as the transport of these compounds across rat intestine. The concentration of d-glucose accumulated or metabolized by the enterocytes (gut wall content) in EJ+MC was higher (p
O.05) dose-dependent inhibition of d-glucose absorption and transport compared to 6, 3 or 1.5 mglmL EJ+MC.
226
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
450 400
--+-- Diebetic control
~
350
__ NonDiabetic Control --+-- Diabetic + EJ 500mg/kg
__ Diabetic + (EJ+MC)
(11300
:g
500mg/kg
g 250 t; -g 200 o
iii 150 100 50
o
Day 0
Day 7
Day14 Days
Day 21
Day 28
Fig 1. Effect of the crude extracts on serum glucose level in alloxan·induced diabetic rats
MC fruit and EJ bark has been found to possess a wide array of bioactive phytochemicals and several classes of biologically active saponins have been isolated (Khanna et al., 1981; Murakami et al., 2001; Matsuur et al., 2002). Bioactive saponins, tannins constituents of natural medicines have been found to suppress the transfer of glucose from the stomach to the small intestine and inhibit glucose and fluid transport at the brush border membrane (Matsuda et al., 2001; Matsuur et al., 2002). Saponins constituents of herbs reduce the permeability barrier to Na+ at enterocytes, thus, discharging the electrochemical gradient and removing the driving force for sugar and fluid transport. Thus, the observed increased inhibitory effect of EJ+MC in the present study might be due to the bioactive compounds such as saponins present in MC fruit (Johnson et al., 1986). The results of the study reveals that EJ bark extract inhibits the uptake of d-glucose, I-tyrosine across rat everted intestinal sacs but when the same drug given in combination with MC shows greater effect by means of increased inhibition of glucose uptake. It is likely that MC fruit and EJ bark possess bioactive phytoconstituents which in combination capable of lowering postprandial blood glucose levels by inhibiting the uptake of glucose across the intestine and at same time increasing metabolism of glucose in cells. As most challenging factor in management of diabetes is to keep PBG as close as to the normal level. Present study supports to corroborate the folklore claim of use ofMC fruits with EJ bark in traditional medicines. In the conclusion, study can be a confirmatory document to claim combination of EJ+MC (50:50) potential alternative drug therapy of, postprandial hyperglycaemia.
Table 1. Effects of graded concentrations of aqueous EJ bark extract (1.5-12 mg/mL) on transport of d-glucose and I-tyrosine across rat everted intestinal sacs Concentration of EJ fruit extract in medium (mg/mL)
d-Glucose transport (MIg tissue wet weight) Mucosal Gut wall disappearance content
0 1.5 3
68.72
±
62.32 49.31
±
6 12
36.44 ± 2.83 40.22 ± 1.80#
±
1. 90 2.06 1.40#
Serosal appearance
15.10
±
1.21
54.10
±
24.37 29.82
±
1.73# 1.23#
40.05 29.63
±
±
24.84 ± 1.36 26.50 ± 0.93#
1.99
2.40# 0.03#,* 15.03 ± 0.86 14.90 ± 1.46# ±
Mucosal disappearance
I-Tyrosine transport (MIg tissue wet weight) Gut wall Serosal appearance content
16.08
±
0.76
1.39
±
0.14
12.91
±
1.42
15.01 12.82
±
1.41 0.83#
1.89 2.07
±
0.30# 0.30#
11.82 9.05
±
0.64 0.56#,*
±
10.03 ± 1.48 10.88 ± 0.68#
±
2.23 ± 0.24 2.09 ± 0.28#
±
8.70 ± 0.66 8.23 ± 0.55#
The results are expressed as means ± S.E.M of seven observations in each group, # p<0.05 from the control without EJ extract added to the mucosal solution. * p<0.05 from those values immediately above (between the different EJ concentrations). Table 2. Effects of graded concentrations of extract MC + EJ (50:50) (1.5-12 mg/mL) on transport of d-glucose and I-tyrosine across rat everted intestinal sacs d-Glucose transport (MIg tissue wet weight) Mucosal Gut wall disappearance content
Serosal appearance
Mucosal disappearance
79.56
±
1.99
13.13
±
2.02
66.43
±
2.12
16.30 ± 0.86
1.32
±
44.86 25.23
±
2.51#
±
±
25.57 29.10 22.22
1.73# 1.18#
41.95
2.08 1.29# 2.58#
±
3 6
70.61 54.33
±
1.36#
19.27
±
0.79# 0.93#
12.82 ± 1.24 10.03 ± 0.54# 09.82 ± 1.27#
1.66 ± 0.30# 1.87 ± 0.41# 2.53 ± 0.33#
1.37 0.42 08.16 ± 0.35# 07.29 ± 0.81#
12
51.03
±
1.87#
26.93
±
0.61#
24.10
±
2.09#
09.09 ± 0.84#
2.00 ± 0.20#
7.09 ± 0.28#
Concentration of EJ fruit extract in medium (mg/mL) 0 1.5
±
±
I-Tyrosine transport (MIg tissue wet weight) Gut wall Serosal appearance content ±
0.22
14.98
±
11.16
±
The results are expressed as means ± S.E.M of seven observations in each group, # p<0.05 from the control without EJ extract added to the mucosal solution. * p<0.05 from those values immediately above (between the different EJ concentrations).
228
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
ACKNOWLEDGEMENTS Authors are indebted to the Prof. R.S. Gaud. Dean, School of Pharmacy and Technology Management, NMIMS University, for provision of all facilities to complete the research work. We are also grateful to Prof. S.B. Gokhale, Principal, SVKM's College of Diploma Pharmacy and Dr. Meena C. HOD Clinical Pharmacology for technical assistance.
REFERENCES Bhattacharya, S.K, Satyan, KS. and Chakrabrati, A. (1997). Effect of Trasina, an Ayurvedic herbal formulation, on pancreatic islet superoxide dismutase activity in hyperglycaemic rats. Ind. J. Exp. Bioi., 35: 297-299. Clarke, E.G.C. and Clarke, M.L. (1977). Veterinary toxicology, 268-277. London: Cassel and Collier Macmilan. Cusi, K and DeFronzo, RA. (1998). Metformin: a review of its metabolic effects. Diabetes Reviews, 6(2): 89-13l. Day, C. (1998). Traditional plant treatments for diabetes mellitus: pharmaceutical foods. Br. J. Nutr., 80: 5-6. Goldner, M. and Gomori, G. (1943). Alloxan induced diabetes. J Endocrinol., 33: 297-9. Gray, A.M. and Flatt, P.R (1998). Actions of traditional antidiabetic plant, Agrimony eupatoria (agrimony): effect on hyperglycemia, cellular glucose metabolism and insulin secretion. Br. J. Nutr., 80: 109-114. Grover J.K, Yadav S. and Vats V. (2002), Medicinal plants of India with anti-diabetic potential. Journal of Ethnopharmacology, 81: 81-100 Johnson, I.T., Jennifer, M., Gee, KP., Caroyn, C. and Fenwick, G.R (1986). Influence of saponins on gut permeability and active nutrient transport in vitro. Journal of Nutrition, 116: 2270-2277. Khanna, P., Jain, S.C., Panagariya, A. and Dixit, V.P. (1981). Hypoglycaemic activity of polypeptide-p from a plant source. Journal Natural Products, 44: 648-655. Kumari, KS. and Devi, KS. (1993). Effect of indigenous drugs on glucuronoglycan metabolism in diabetic hypertensive rabbits. Ind. J. Exp. Biol., 31: 595-599. Lin, C.C. (1992). Crude drugs for the treatment of diabetes mellitus in Taiwan. Am. J. Chin. Med., 20(3-4): 269-279. Liu, RY. and Wang, G.Q. (1996). A survey on drugs synthesized for antidiabetes. Journal of Shenyang Pharmaceutical Unwersity, 13: 148-153. Lowry, O.H., Rosebrough, N.J., FaIT, A.L. and Randall, RJ. (1951). Protein measurement with the folin phenol reagent. J. Bioi. Chem., 193: 269-275. Lowry, O.H., Rosenbrough, N.J., FaIT, A.L. and Randall, RJ. (1951). Protein measurements with the Folin phenol reagent. Journal of Biological Chemistry, 193: 265-275. Matsuda, H., Murakami, T., Li, Y., Matsumura, N., Yam ahara, J. and Yoshikawa, M. (1998). Antidiabetic principle of natural medicines. III. Structure-related inhibitory activity and action mode of oleanolic acid glycosides on hypoglycemic activity. Chemical Pharmaceutical Bulletin, 46: 1399-1403. Matsuur, H., Asakawa, C., Kurimoto, M. and Mizutani, J. (2002). Alpha-glucosidase inhibitor from the seeds of balsam pear (Momord!ca charantia) and the fruit bodies of Grifola frondosa. Bioscience Biotechnology Biochemistry, 66: 1576-578. Mitra, S.K, Gopumadhavan, S., Muralidhar, T.S., Anturlikar, S.D. and Sujatha, M.B. (1996). Effect of a herbomineral preparation D-400 in streptozotoc in induced diabetic rats. J. Ethnopharmacoi., 54: 41-46. Murakami, T., Emoto, A., Matsuda, H. and Yoshikawa, M. (2001). Medicinal foodstuffs. XXI. Structures of new cucurbitane-type triterpene glycosides, goyaglycosides-a, -b, -c, -d, -e, f, -g and -h, and new oleanane-type triterpene saponins, goyaglycosides I, II and III, from the fresh fruit of Japanese Momordica charantia L. Chemical Pharmaceutical Bulletin, 49: 54-63.
E. jambolana and M. charantia in Respect to Safety and Antidiabetic Effect
229
Nadkarni, A.K. (1954). Indian Materia Medica, Popular Prakashan, Bombay, 1221. Ospinag, L.F. and Serrano, RP. (1995). Plants used as antidiabetics in Colombian folk medicine. Rev. Colomb. Clenc. Quimico·Farm., 23: 81-94. Reaven, G.M. (1983). Effect of age and diet on insulin secretion and insulin action in the rat. Am. J. Med., 74: 69. Sharma, R and Misra, A. (1993), Autoimmunity and gestational diabetes. The National Medlcal Journal of Indw, 6: 272-273. Shimada, H., Kawamori, S. and Kawahara, Y. (1998). Effect of an aqueous extracts of Salacia reticulata, a useful plant in Srilanka, on postprandial hyperglycemia in rats and human. Nippon Eiyo Shakuryo Gakkaiski., 51: 279-287. Silva, F.RM.B., Szpoganicz, B., Pizzolati, M.G., Willrich, M.A.V. and Souza, E. (2002). Acute effect of Bauhinia forficata on serum glucose levels in normal and alloxan-induced diabetic rats. Journal of Ethnopharmacology, 83: 33-37. Trinder, P. (1976). In: Varley, H., Gowenlock, A.H. and Bell, M. (Eds.). Practical Clinical Biochemistry. William Heinemann Medicinal Books Ltd., London, p. 389 Zhao, S.Y. (1999). Drugs to treat Diabetes. Guowai Yiyao-Hechengyao, Shenghuayao, Zhiji, 20: 130-135.
"This page is Intentionally Left Blank"
10 Larvicidal and Antimicrobial Activities of Seeds of Annona cornifolia A. St. -Hil. (Annonaceae) LUCIANA ALVES R.S. LIMA\ MARIAAMELIAD. BOAVENTURA\ JACQUELINE A. TAKAHASHI l AND LUCIA P.S. PIMENTA l
*
ABSTRACT The hexane and ethanol extracts from Annona cornifolia A. St. -Hil seeds showed expressive activity in Artemia salina (brine shrimp) and antimicrobial tests. A fraction from hexane extract rich in fatty acids and the corresponding fatty acid methyl esters fraction showed antibacterial and antifungal activities. Methyl esters were prepared from saponifiable fraction of hexane extract with methanol/sulfuric acid and were identified by GC and GC-MS. A fraction obtained from chloroform extract, positive for acetogenins, was extremely active against Candida albicans. Key words : Annona cornifolia, Annonaceae, antimicrobial activity, fatty acid methyl esters, larvicidal activity
INTRODUCTION Plants from Annonaceae family are very important sources of edible fruits and they are used in folk medicine as antitumor, parasiticide, insecticide, antifever, antidysentery, antiedema (Cepleanu et al., 1993), anti-influenza, anti-insomnia (Wang et al., 2002) and antidiarrhoea agents (Correa et al., 1984; Leboeuf et al., 1982). Annona is far the greater genus in this family and species such as A. crassiflora, A. squamosa and A. muricata have a wide use in traditional medicine against diabetes, malaria and infections (Sousa et al., 1991). These activities, from the phytochemical viewpoint, were initially associated only with the isolation of alkaloids, very common 1. Departamento de Quimica, Instituto de Ciencias Exatas, UFMG, Av. Antonio Carlos 6627, Belo Horizonte, MG, CEP.: 31270-901, Brazil. * Corresponding author: E-mail: [email protected]
232
Comp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
phytochemical constituents of this family. Nowadays, several activities associated to other classes of natural compounds isolated from this family, such as annonaceous acetogenins, have been reported (Alali et al., 1999). Annona cornifolia A. St. -Hil. is native from southeast of Brazil and its green fruits are popularly used to heal chronic ulcers (Correa, 1984). The only phytochemical study of this species, to our knowledge, has been done by our group and the isolation and characterization of three acetogenin were described (Santos et al., 2006; Santos et al., 2007).
Since most of the activities related to Annonaceae plants, including Annona species, concern to their anti-infectious potential, A. cornifolia was screened by larvicidal (Brine Shrimp Lethality test) and antibacterial! antifungal protocols.
MATERIALS AND METHODS General
IR spectra were recorded on a ShimadzulIR-408 spectrophotometer. Fatty acids methyl esters analysis was carried out with a Varian CP-3380 Gas Chromatograph using hydrogen as the carrier gas. A GE Science BPI column (12 m x 0.25 mm, 100% dimetylpolysiloxane, 0.25 mm) was used. The temperature of the GC oven was held at 150°C and programmed to 240°C with increase of lOoC/min; injection temperature was 250°C and detector, temperature 260°C. Split ratio was 1/80. Analysis GC-MS were performed in a Hewlett Packard 5890 Serie II Chromatograph coupled in a Hewlett Packard 5989A mass spectrometer, using electron ionization. Oven flow was 2 mUmin and mass range was 40-400 m/z. The fatty acids methyl esters were identified by comparison of their retention times with those of standards and by using the Wiley 138 Library database search. Plant Collection, Identification and Extract Preparation
Fruits of Annona cornifolia A. St. -Hil. were collected in Curvelo city region, Minas Gerais State, Brazil, from January to March 1998. The species was identified by Dr. R. Mello-Silva and a voucher specimen (BHCB 68114) was deposited at the Instituto de Ciencias Biologicas Herbarium, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil. The fruits were open, the seeds removed, washed, dried at 40°C, powdered (850.0 g) and extracted exhaustively with hexane to give extract F01 (79.5 g) and then with ethanol to give extract F02 (120.0 g), after solvent removal. The latter was dissolved in methanol!water (7:3) and extracted with chloroform to give F03 (7.3 g). Hexane extract (F01) was fractionated by column chromatography on silica gel using gradient systems of n-hexane/CH 2 Cl/AcOEtlCH3 0H in increasing polarities; a fraction rich in fatty acids (F01-1, 53.2 g) was isolated. Part of this fraction (500 mg)
Larvicidal and Antimicrobial Activities of Seeds of Annona cornifolia
233
was refluxed with 1.0 mollL methanolic sodium hydroxide solution for 30 min. and then extracted with ethyl ether. The aqueous phase was acidified with 1.0 mollL HCI solution and extracted with ethyl ether. The organic phase was dissolved in hexane and then refluxed with H 2S0 4 methanolic solution 2% v/v for 1 h. 370 mg of a mixture of fatty acid methyl esters (F01-1Me) were obtained after extraction and solvent elimination. Part of chloroform fraction (F03, 1.0 g) was submitted to medium pressure liquid chromatography using hexane/toluenlAcOEtlEtOHlMeOH in increasing polarities, to furnish several fractions, that were tested for acetogenins by TLC. Acetogenins containing fractions were expected to show pink color by Kedde's reagent and orange color by Dragendorffs reagent. Infrared profile were also obtained. Only fraction F03-7 was positive for acetogenins. Afterwards acetogenins were isolated from this fraction and were related elsewhere (Santos et al., 2006; Santos et al., 2007; Lima et al., 2008).
Larvicidal Bioassay The methodology used was described by Pimenta et al. (2003) and Stefanello et al. (2006). Artemia salina encysted eggs (10 mg) were incubated in 100 mL of seawater under artificial light at 28°C and pH 7-8. After incubation for 24 h, nauplii were collected with a Pasteur pipette and kept for additional 24 h under the same conditions to reach the metanauplii stage. The samples (triplicate) to be assayed were dissolved in dimethylsulfoxide (DMSO) (2 mg/400 ilL or 2 mg/1000 ilL) and diluted serially (10, 20, 30, 50 IlU5 mL) in seawater. About 10-20 nauplii were added to each set of tubes containing the samples. Controls with 50 ilL of DMSO in seawater were included in each experiment. Lapachol dissolved in DMSO was used as positive control. 24 h later, the number of survivors was counted, recorded and the lethal concentration 50% (LC 5o value) and 95% confidence intervals were calculated by probit analysis (Finney, 1974).
Antimicrobial Bioassay The methodology used was described by Miguel et al. (1996); Bisagno et al. (2000); Almeida et al. (2006) and Smania et al. (2007). Staphylococcus aureus (ATCC 25985), Streptococcus pyogenes (ATCC 19615), both gram positive, Salmonella typhimurium (ATCC 13311), gram negative and the yeast Candida albicans were used for this test. The minimum inhibitory concentration (MIC) was evaluated by the macro dilution test using standard inoculums of 10-5 CFU mL-l. Serial dilutions of the test compounds, previously dissolved in DMSO, were prepared to final concentrations of 550.00, 275.00, 137.50, 68.75, 34.38, 17.18, 8.59, 4.30, 2.15 and 1.07 llg/mL. To each tube were added 100 ilL of a 24 h old inoculum. The MIC, defined as the lowest concentration of the test compound, which inhibits the visible growth after 18 h, was determined visually after
234
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
incubation for 18 h at 37°C. Tests using DMSO as negative control and chloramphenicol and ketoconazol as positive controls were carried out in paralleL All tests were performed in duplicate with full agreement between both results.
RESULTS AND DISCUSSION Phytochemical analysis of extracts and fractions obtained from A. cornifolia seeds led to the identification of a fraction positive for acetogenins (F03-7), obtained from chloroform extract (F03). Both F03 and F03-7, as well as F01 (hexane extract), F01-1 (fatty acids fraction), F01-1Me (fatty acid methyl esters fraction) and F02 (ethanol extract) were tested for cytotoxicity by brine shrimp lethality (A. salina) test. The results obtained from this bioassay are shown in Table 1. LD50 values, with 95% confidence range, in IJ,gimL, were determined using probits method (Finney, 1974). Hexane extract (F01), fraction rich in fatty acid (F01-l), and fatty acid methyl ester fraction (F01-1Me) showed good cytotoxic activity (LD 50 = 10.00, 9.60 and 8.77 IJ,g/mL, respectively), even higher than the activity presented by the standard, lapachol LD50 = 70 mgimL (61 < LD50 < 81). Fraction F01-1Me was analysed by gas chromatography and the following fatty acid methyl esters were identified: miristate (C-14); palmitate (C-16); stearate (C-18); oleate (C-18, ~9); linoleate (C-18, ~9,12) and linolenate (C-18, ~9,12,15), in 0.17, 16.92, 5.56, 51.46, 19.12 and 0.81%, respectively. Table 1. Larvicidal (brine shrimp lethality bioassay) and antimicrobial (minimum inhibitory concentration, MIG) activities for extracts and fractions from seeds of Annona cornifolia Extracts and fractions
FOl F02 F01-1 F01-1Me F03 F03-7 Lapachol b Ghloranphenicol Ketoconazol d
Larvicidal Minimum inhibitory concentration (mg/mL) (MIe) activity Salmonella StaphyloccoccusStreptococcus Candida aureus pyogenes albicans typhimurium LDso a(Wl/mL) 10.00 0.20 9.60 8.77 0.13 0.16 68.09 C
137.50 137.50 275.00 550.00 137.50
68.75 137.50 137.50 275.00 137.50
137.50 137.50 137.50 275.00 137.50
8.40
33.60
4.20
34.38 68.75 68.75 nd 2.15
40.60
a Dosis that kills 50% ofthe individuals of a population; b control used in the brine shrimp larvicidal bioassay; C control used in the MIG test for S. aureus, S. pyogenes, Salmonella typhimurium; d control used in the MIG test for C. albicans; nd = not detected.
Larvicidal and Antimicrobial Activities of Seeds of Annona cornifolia
235
The saponifiable part of the fraction F01-1 showed 6 fatty acids identified as methyl ester. The total of unsaturated fatty acids methyl esters found in F01-1Me was 71.39% (Table 2). Polyunsaturated fatty acids, when screened by the brine shrimp A. salina test, are reported to have higher toxicity compared to satured fatty acids (lkawa, 2004). According to this report, the toxic activity of such fatty acids hypothetically arises from the corresponding oxidation products, derived from photooxidation or metabolic processes. Table 2. Fatty acid composition ofthe FOl-l hexane fraction of A. cornifolia seeds in its methyl ester form Compund ~ethylmyristate ~ethyl
palmitate ~ethyl stearate ~ethyl oleate ~ethyllinoleate ~ethyllinolenate
Relative percentage 0.17 16.92 5.56 51.46 19.12 0.81
F02, F03 and F03-7 were highly active (LD 50 = 0.20 llg/mL, 0.13 and 0.161lg/mL, respectively), in brine shrimp CA. salina) test, presenting results compatible with literature values found for mixtures of acetogenins (Pimenta et al., 2003). All fractions were also tested for their antimicrobial activity. Minimum inhibitory concentration (MIC) was determined by macrodilution method for ten test concentrations ranging from 550 to 1.07 /lWmL. MIC values were determined after incubation at 37°C, for 14 h. All extracts and fractions were active (Table 1). F03-7 was not tested, due the little amount available. F01, constituted by a mixture of fatty acids, showed activity, against S. aureus, close to the control value. It is already known that even traces of certain long chain fatty acids inhibit microorganisms growth, specially Gram-positive bacteria, by affecting cell permeability and transport of nutrients (Wang & Johnson, 1992). Fatty acid monoesters of glycerol have strong bacteriostatic activity against B. subtilis and S. aureus (Kato & Shibasaki, 1975). According to Kabara (1987), esterification of polyhydroxyl compounds, as glycerol, with fatty acids generally leads to more active derivatives, whereas the same reaction with monohydroxyl compound cause deactivation of the fatty acid. In agreement with this information, esterified fraction (F01-1Me) showed less activity than the original fraction containing the fatty acids. F03, rich in acetogenins, showed good activity against Salmonella typhimurium, Staphylococcus aureus and Streptococcus pyogenes. Acetogenins isolated from Uvaria hookeri and Uvaria narum are reported to possess antibacterial and antifungal activities (Padmaja et al., 1993).
236
Camp. Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
The present study shows that traditional use of A. cornifolia fruits, such as against ulcers, can be associated to the cytotoxic and antibacterial activities found in their extracts. A. cornifolia seems to be extremely interesting as source of novel antifungal agents. Both hexane and ethanol extracts (F01 and F02) were highly active against Candida albicans, F01 being even more active than the control used (ketoconazol). F03, the chloroform fraction obtained from F02, presented a MIC = 2.15 ~g/mL, being almost 20 times more active than the control. From the fraction F03 five acetogenins were isolated that have showed great activity in Brine Shrimp Lethality test (Santos et al., 2006; Santos et al., 2007; Pimenta et al., 2008).
AKNOWLEDGEMENTS The authors thank Conselho Nacional de Desenvolvimento Cientifico e Tecnol6gico (CNPq) for LARSL, MADB and JAT grants. The authors also thank FAPEMIG for financial support (grant CDS-853106).
REFERENCES Alali, F.Q., Liu X.-X. and McLaughlin, J.L. (1999). Annonaceous acetogenins: recent progress. J Nat Prod., 62: 504-540. Almeida, J.RG.S., Filho RN.S., Nunes, X.P., Dias, C.S., Pereira, F.O. and Lima, E.O. (2006). Antimicrobial activity of the essential oil of Bowdichia virgilioides Kunt. Rev Bras Farmacogn., 16: 638-641. Bisagno, G., Sanogo, R, Marino, A., D'Angelo, V., Germano, M.P., De Pasquale, R and Pizza C. (2000). Antimicrobial activity of Mitracarpus scaber extract and isolated constituents. Letters in Appl Microbiol., 30: 105-108. Cepleanu, F., Ohtani, K., Hamburger, M., Gupta, M.P., Solis, P. and Hostettmann, K. (1993). Novel acetogenins from leaves of A. purpurea. Helv Chim Acta, 76: 1379-1388. Correa, M.P. (1984). Dicionario de plantas uteis do Brasil e de plantas ex6ticas cultivadas. Rio de Janeiro: Imprensa Nacional, v. 1, pp. 151-162. Finney, D.J. (1974). Probit analysis, a statical treatment of sigmoid response curve. 3 ed. Cambrige: University Press, pp. 76-80. Ikawa, M. (2004). Algal polyunsaturated fatty acids and effects on plankton ecology and other organisms. UNH Center for Freshwater Biology Research, 6: 17-44. Kabara, J.J. (1987). Fatty acids esters as antimicrobiallinseticidal agents. In: ACS Symposium Series 325 (Ecol. Metab. Plant Lipids): 220-238. Kato, N. and Shibasaki, 1. (1975). Comparison of antimicrobial activities of fatty acids and their esters. J. Ferment Technol., 53: 793-801. Leboeuf, M., Cave A., Braumik, P.K., Mukherjee, B.E. and Mukherjee, R (1982). The phytochemistry of the annonaceae. Phytochemistry, 21: 2783-2813. Miguel, O.G., Lima, E.O., Morais, V.M.F., Gomes, S.T.A., Monache, F.D., Bella-Cruz, A., Bella-Cruz, RC. and Cechinel, F.v. (1996). Antimicrobial activity of constituents isolated from Lichnophora sal~cifolia (Asteraceae). Phytoter Res., 10: 694-696. Padmaja, D., Thankamany, V. and Hisham, A. (1993). Antibacterial, antifungal and antihelmintic activities of root barks of Uvaria hookeri and Uvaria narum. J Ethnopharmacol, 40: 181-186. Pimenta, L.P.S., Pinto, GB, Takahashi, J.A., Silva, L.G.F. and Boaventura, M.A.D. (2003). Biological screening of annonaceous Brazilian medicinal using Artemia salina (brine shrimp test). Phytomedicine, 10: 209-212.
Larvicidal and Antimicrobial Activities of Seeds of Annona comifolia
237
Pimenta, L.P.S., Lima, L.A.R. dos S. and Boaventura, M.A.D. (2008). Submitted for publication. Santos, L.A.R., Boaventura, M.A.D. and Pimenta, L.P.S. (2006). Cornifolin, a new bistetrahydrofuran annonaceous acetogenin from Annona cornifolia. Biochem System Ecol., 34: 78-82. Santos, L.A.R., Boaventura, M.A.D. and Pimenta, L.P.S. (2007). Acetogeninas de Anonaceas Bioativas isoladas das sementes de Annona cornifolia A. St.-Hill. Rev. Bras. PI. Med., 9: 48-5l. Smania, E.F.A., Monache, F.D., Yunes, R.A., Paulert, R. and Junior, A.S. (2007). Antimicrobial activity of methyl australate from Ganoderma australe. Rev Bras Farmacogn., 17: 14-16. Sousa, M.P., Matos, M.E.O. and Matos, F.J.A. (1991). Constituintes quimicos de plantas medicinais brasileiras. Fortaleza: Impr. UniversitarialUFC. Stefanello, M.E.A., Salvador, M.J., Ito, LY. and Macari, P.A.T. (2006). Avalia~i'i.o da atividade antimicrobiana e citot6xica de extratos de Gochnatia polymorpha ssp floccosa. Rev Bras Farmacogn., 16: 525-530. Wang, L.L. and Johnson, E.A. (1992). Inhibition of Listeria monocytogenes by fatty acids and monoglycerides. Appl Environ Microbwl., 58: 624-629. Wang, L.Q., Min, B.-S, Li, Y., Nakamura, N., Qin, G.-W., Li, C.-J. and Hattori, M. (2002). Annonaceous acetogenins from the leaves of Annona montana. Bioorg Med Chem., 10: 561-565.
"This page is Intentionally Left Blank"
11 Antidepressant Activity of the Extracts of Three Hypericum species Native to China DEPO YANG!, JrE BAI l ,2, ZHEN Lr!, GEORGE
Q L r3 AND DONGMEr WANG l ,*
ABSTRACT
The objective of this study was to evaluate three Chinese species ofHypericum genus (Hypericaceae): Hypericum erectum subsp. longisepalum, H. hubeiense and H. seniavinii, as the new sources of antidepressant phytomedicine. The hydroalcoholic extracts of the three Hypericum plants were evaluated for their antidepressant effects by forced swimming test (FST) and tail suspension test (TST) in mice. The mechanisms of action were also investigated by 5-HT-induced head twitches, potentiation of NA toxicity, and elevated plus-maze in mice. All the three extracts (at the doses of250 or 500 mg/kg, or both) significantly shortened the immobility time in FST and TST. These extracts potentiated the head twitches induced by 5-HT, and H. hubeiense extract (at the dose of 500 mg/kg) also showed an increase of noradrenaline toxicity. In addition, H. erectum and H. hubeiense extracts showed anxiolytic action in the elevated plus-maze test. The results suggest that these three Hypericum extracts possess antidepressant activity, which may be mediated by serotonergic system, and noradrenergic system may also be involved for H. hubeiense. The three Chinese Hypericum species possess activity similar to H. perforatum and have the potential to be used as new sources of antidepressant products. Key words : Antidepressant, forced swimming test, hypericum, tail suspension test 1. School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510 080, P. R.
China. 2. Chemistry Department, Tsinghua University, Beijing 100 084, P.R. China. 3. Herbal Medicines Research and Education Centre, Faculty of Pharmacy, The University of Sydney, NSW 2006, Australia. * Corresponding author : E-mail: lsswdm®mail.sysu.edu.cn.
240
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
INTRODUCTION Hypericum perforatum L. (St. John's wort) is herbaceous plant used for the treatment ofthe mild-to-moderate depression (Linde et al., 1996; Kasper et al., 2001). It is used in traditional Chinese medicine for the treatment of burns, bruises, swelling, inflammation, and skin diseases (Wu et al., 2002). It has been demonstrated that hypericins, hyperforin and flavonoids are the main active components (Jiang et al., 2004; Sanchez-Mateo et al., 2005). Recent reports have raised the concerns on its interference effect on the metabolism of some pharmaceutical drugs which may limit its future application for some patients (Jiang et al., 2004). Therefore, there is a need to improve the safety profile of St. John's wort and extracts from other species of Hypericum which may offer the alternative (Sanchez-Mateo et al., 2005). The current study aims to identify new source of Hypericum of antidepressant activity from Chinese Hypericum species. There are over 60 Hypericum species distributing in China, but only a few of them have been studied for their chemical constituents and pharmacological activities (Wu et al., 2002; Fu et al., 2006). Recently new species of Hypericum were collected and identified by our laboratory (Wu et al., 2004). In the current study, we reported the antidepressant activity and the possible mechanisms of three Chinese Hypericum: Hypericum erectum subsp. longisepalum L. H. Wu (H. erectum), H. hubeiense L. H. Wu et D. P. Yang, and H. seniavinii Maxim, using five well established animal models.
MATERIALS AND METHODS Plant Materials The aerial parts of H. erectum, H. hubeiense and H. seniavinii were collected in Badong (Hubei Province, China) during the flowering period in August 2001, and dried at room temperature avoiding direct sunlight. The voucher specimens (No. 01020, No. 01035, No. 01034, respectively) were deposited at Biological Museum of Sun Yat-sen University, Guangzhou, China. The aerial parts of these plant samples were powdered for extraction. The hydroalcoholic extracts were obtained by maceration of the powdered samples with 6-fold (w/v) amount of 90% aqueous ethanol for overnight at room temperature, and this procedure was repeated twice. The extracts were filtered and dried under reduced pressure below 45°C. The contents of hypericins (hypericin and pseudohypericin) were determined by HPLC and the total flavonoids determined by UV using published methods (Zheng et al., 2003).
Antidepressant Activity of the Extracts of Hypericum Species
241
Drugs The following drugs were used as control in the study: imipramine hydrochloride (Jiufu, China), amitriptyline hydrochloride (Shizhou, China), 5-hydroxytryptophan (Acros, USA), N-methyl-N-propargyl-benzylamine (Pargyline) (Acros, USA), norepinephrine (Changji, China), and diazepam (Siyao, China).
Animals and Drug Administration NIH male mice (18-22 g) were provided by the Medical Animal Center, Guangzhou University of Chinese Medicine (China). The naive animals were kept in a 12 h light/dark cycle (light on started at 07:00) at ambient temperature of 25 ± 1°C. Plant extracts and control drugs were suspended in a 0.5% carboxymethyl cellulose aqueous solution (vehicle) and adjusted to various concentrations so that the final volume given orally was 15 mU kg, and the final dosage in mice were 250, and 500 mglkg body weight. They were given orally by gavage 1 h before the tests. Control animals received vehicle under the same conditions. Behavioral observations took place between 8:00 and 15:00 o'clock. All studies were performed in accordance with the Experimental Animal Management Regulations of National Science and Technology Commission, China (November 14th 1988, Article No.2). In each experimental paradigm, the observers were blind of the treatment protocol. Imipramine and amitriptyline (20 mg/kg) were given by intraperitoneal route (i.p.) 10 min before FST, and TST respectively. In the elevated plusmaze test, diazepam (3 mglkg) or vehicle was given orally 1 h before the tests were performed. Experiments were conducted between 8:00-15:00 o'clock. The mice were used just for one time. Each group of behavioral experiments consisted of 12 mice, unless otherwise stated.
Preliminary Acute Toxicity Limit test was performed to observe the acute toxicity of the three Hypericum extracts. There were six mice in each group. Animals received decreasing doses of H. erectum, H. hubeiense and H. seniavinii extracts with the initial dose of 5 glkg (p.o.), and were placed in empty cages to observe signs of toxicity. Observation was carried out until 7 days after administration.
Forced Swimming Test (FST) in Mice The forced swimming test was performed according to the methods described by Porsolt et al. (1977). Mice were individually placed in a cylindrical glass swim vessel (25 x 12 cm i.d.) filled with 12 cm of water maintaining at 23-25°C. The total immobility time was observed during the last 4 min of a single 6 min test session by unaided eyes of a trained observer, who was blind to the experimental conditions. Mice were
242
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
considered immobile when they showed no further attempts to escape except the necessary movements to keep their heads above the water.
Tail Suspension Test (TST) in Mice After 1 h following withdrawal, mice were individually taken out from the vivarium to an adjoining room and immediately suspended by the tail to a horizontal ring-stand bar (The distance from the Table top was 60 cm) using adhesive tape affixed 1 cm from the tip of the tail. The testing room was brightly lit. The mouse was 15 cm away from the nearest object and both acoustically and visually isolated. The cumulative period of immobility during an observation period of 6 min was recorded by a video camera. The video tapes were scored by a trained observer who was blind to the experimental conditions. Mice were considered immobile only when they hung passively and were completely motionless (Steru et al., 1985).
5-HT-induced Head Switches in Mice Test drugs were orally administered 30 min before 100 mg/kg i.p. pargyline, and then 1 h later, i.p. 20 mg/kg 5-HT. Mter administration, the mice were placed in empty cages and the number of head twitches was counted during 15-35 min after 5-HT injection (Goodwin et al., 1984).
Potentiation ofNA Toxicity in Mice Eight groups of mice, ten in each group, were injected NA (3 mglkg) subcutaneously 60 min after being administrated the extracts (250 and 500 mg/kg, p.o.), vehicle (p.o.), and amitriptyline (20 mg/kg, p.o.), respectively. The number of dead mice was counted 48 h after the administration of test drug (Alpermann et al., 1992).
Elevated Plus-Maze in Mice The mouse plus-maze consisted of two opposite open arms (30 x 5 cm) crossed with two closed arms of the same dimension with 15 cm height. The arms were connected with a central square (5 x 5 cm) elevated 45 cm above floor level. The animal was placed individually in the center ofthe maze facing a closed arm. The number of entries into open and closed arms, and the time spent in each arm during the next 5 min were evaluated by an observer inside the room (Willner et al., 1984; Lister et al., 1987). During the experiment, when each test of 5 min was completed, the maze was carefully cleaned with 20% ethanol solution and then thoroughly dried.
Statistical Analysis Data analysis was performed by one-way analysis of variance (ANOVA) with the Dunnett post-hoc test for multiple comparisons by SPSS 10.0 software. All data were expressed as means ± S.E.M. and the level of statistical significance was set at p<0.05.
Antidepressant Activity of the Extracts of Hypericum Species
243
RESULTS Preliminary Acute Toxicity In limit test, H. hubeiense extract did not cause any toxicity manifestations until a dose of 5 g/kg (p.o.), while H. erectum and H. seniavinii extracts induced mortality at lower doses. H. seniavinii and H. erectum extracts showed no toxicity with doses up to 3.5 g/kg, and 1.75 g/kg, respectively. It was observed that the body weight of the mice treated with H. seniavinii extract was significantly lower than those of control group 3 days after administration at a dose of 3.5 g/kg, and then recovered to normal body weight 7 days later. No body weight reduction was observed in H. erectum or H. hubeiense treated groups.
Behavioral Despair Test The activities of the three Hypericum extracts on the forced swimming test are shown in Fig 1. The tricyclic antidepressant imipramine (20 mg! kg) reduced the immobility time significantly (69.4 ± 8.5 seconds from control 105.9 ± 19.2 seconds, p<0.05). At 250 and 500 mg/kg, both H. hubeiense and H. erectum extracts, shortened the immobility time of mice significantly in FST (p<0.05 or p
~ (/J
~
~
r:J 250
140 120 100 80
mg/kg
!iJ 500
mg/kg
*
.c ~ 60 E 40 20
o
Vehicle
Imipramine
Fig 1. Effect of the extracts of three Hypericum species on the total duration of immobility in the forced swimming test (FST) in mice. Results are means ± S.E.M., n=12, *p
The activities of the three Hypericum extracts on the tail suspension test are shown in Fig 2. The tricyclic antidepressant imipramine (20 mg! kg) reduced the immobility time significantly (43.2 ± 10.6 seconds from control 107.9 ± 10.4 seconds, p<0.05). H. erectum extract shortened the immobility time not significantly at 250 mg/kg (86.0 ± 16.4 s), and significantly at 500 mg/kg (73.0 ± 8.9 s). H. hubeiense extract shortened the immobility time significantly at 250 mg/kg (74.2 ± 9.0 s), but not
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
244
significantly at 500 mg/kg (78.5 ± 12.5 s). H. seniavinii at 250 and 500 mg/ kg shortened the immobility time significantly (71.7 ± 11.2, 53.0 ± 9.3 s respectively). 140
D 2S0mg/kg
ISlSOOmg/kg
::§: 120 ~ 100 :;:::; 80
~ :c
60 o E 40 E 20
o
L -_ _~-L~~~_ _~~L-L-~~~~______-L~
Vehicle
HE
HH
HS
Imipramine
Fig 2. Effects ofthe extracts ofthree Hypericum species on the total duration of immobility in the tail suspension test (TST) in mice. Results are means ± S.E.M., n=12, *p<0.05, **p
5-HT - Induced Head Twitches in Mice In order to understand the mechanism of actions of antidepressant effects, the 5-HT - induced head twitches and potentiation of NA toxicity models were performed. The effects of the extracts of three Hypericum species on 5-HT induced head twitches are shown in Table 1, the extracts of H. erectum (250 and 500 mg/kg), H. hubeiense (250 mg/kg) and H. seniavinii (500 mg/ kg) produced significant potentiation of the head twitches, while imipramine did not produce significant effect at the dose of 20 mg/kg. In the control group there was no mice died, but in all the extract groups and imipramine group, the mice died at different death rates. The number of head twitches was counted from the survival mice. Table 1. Effects of the extracts of three Hypericum species on 5-HT induced head twitches (means ± S.E.M) Treatment Vehicle H. erectum H. seniavinii H. hubeiense
Imipramine
Dose (mglkg)
250 500 250 500 250 500 20
*p<0.05, **p
No. of head twitches 2.0 30.7 10.8 3.8 7.1 6.9 2.5 3.3
± ± ± ± ± ± ± ±
0.4 10.1** 0.7** 1.9 0.8* 0.7* 0.9 1.8
Rate of death 0/12 4/10 1110 3/10 2.110 1110 2.110 2.110
Antidepressant Activity of the Extracts of Hypericum Species
245
Potentiation ofNA Toxicity No death of mice was observed in control group at the sub-lethal dose (3 mglkg, sc.) ofNA. The results, as shown in Table 2, demonstrated that only H. hubeiense extract (500 mglkg) produced the potentiation of norepinephrine toxicity, similar to the control amitriptyline, whereas H. seniauini and H. erectum (250 and 500 mglkg) did not have the potentiation effect after acute administration. Table 2. Potentiation ofNA (3 mg/kg, sc.) toxicity in mice (means ± S.E.M, n=12) Dose (mg/kg)
Treatment Vehicle H. erectum
Rate of death
o o o o o o
250 500 250 500 250 500 20
H. seniavinii H. hubeiense Amitriptyline
3/12** 5/12**
*p<0.05, **p
Elevated Plus-Maze The results of the anxiolytic activity of the extracts were shown in Table 3. H. erectum, H. hubeiense extracts (250 and 500 mglkg) and the anxiolytic benzodiazepine drug, diazepam significantly increased residence time of animals in open arms and the number of entries to open arms. At the dosage Table 3. Effects of the extracts of three Hypericum species in the elevated plus-maze (EPM) test in mice (means ± S.E.M, n=12) Open arms Treatment
Dose (mg/kg)
Entries (N)
Vehicle H. erectum
H. seniavinii H. hubeiense Diazepam
250 500 250 500 250 500 3
5.2 8.7 11.6 7.0 6.8 10.2 9.3 9.6
± 0.7 ± 1.3* ± 3.1** ± 3.4 ± 1.8 ± 2.1** ± 1.3* ± 2.0*
*p<0.05, **p
Close arms
Time spent (s) 27.2 40.7 43.3 35.8 35.7 67.7 40.3 62.5
± 5.6 ± 12.3* ± 19.1* ± 6.9 ± 7.0 ± 16.6** ± 18.9* ± 11.0**
Entries (N)
11.3 12.8 11.4 9.7 14.8 10.1 10.2 12.8
± 2.1 ± 3.1 ± 3.7 ± 2.1 ± 3.1 ± 2.5 ± 4.1 ± 2.6
Time spent (s) 233.5 199.2 128.0 190.6 206.7 130.7 129.9 133.1
± 15.8 ± 19.5 ± 13.0** ± 20.2 ± 34.1 ± 10.6**
9.5** ± 8.0** ±
246
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
of 500 mg/kg the two extracts decreased residence time in enclosed arms significantly. However, there was no significant change observed in the number of entries to the closed arm for the control and all extracts. The other species H. seniavinii extract (250 and 500 mg/kg) did not significantly affect the residence time and the number of entries in open arms and closed arms.
DISCUSSION The forced swimming test (FST) (Porsolt et al., 1977) and the tail suspension test (TST) (Steru et al., 1985) are the typical behavioral models commonly used in rodent for screening the antidepressant drugs. The immobility displayed in rodents, subjected to an unavoidable and inescapable stress, is known to reflect the behavioral despair, which may in turn reflect depressive disorders in humans (Goodwin et al., 1984; Alpermann et al., 1992). We studied the hydroalcoholic extracts of three Hypericum species endemic to China using these behavioral despair test models in mice. The results indicated that all the extracts (at the doses of 250 or 500 mg/kg, or both), and the conventional TCA antidepressants imipramine and amitriptyline, reduced the immobility time significantly. Serotonin and norepinephrine are known to play key roles in depression occurrences. Drugs that inhibit the reuptake of serotonin and norepinephrine are known to be effective in the treatment of major depression. H. hubeiense, H. seniavinii and H. erectum extracts could all significantly potentiate the 5-HT-induced head twitches compared with the control group. Additionally, all the extracts and imipramine induced partial mice death at different rates, acompanying with simultaneously drooling and frequent head twitches. These results indicate the possible actions of 5-HT reuptake inhibition (Shank et al., 1987) and potentiation of 5-HT toxicity in the serotoninergic system. It has been reported that some antidepressant drugs, such as, fiuoxetine, dexfenfluramine, may cause weight loss, along with their activities in increasing 5-HT availability in the synaptic clefts (McTavish et al., 1992; Goldstein et al., 1994). The weightlosing effect of H. seniavinii extract was observed in Limit Test, which also indicates the possibility of serotoninergic system involvement. The potentiation of norepinephrine toxicity in mice was examined to investigate the performance of the extracts in the noradrenergic system. In the experiment, 50% of the animals treated with amitriptyline died. However, only H. hubeiense extract could potentiate the norepinephrine toxicity at the dose of 500 mg/kg with 30% induced-mortality. H. erectum and H. seniavinii extracts showed no action in the noradrenergic system at the tested doses. No mortality occurred in the control group, neither. It suggested that the antidepressant activity of H. hubeiense extract maybe related to its possible effect in noradrenergic uptake inhibition.
Antidepressant Activity of the Extracts of Hypericum Species
247
Human depression is often accompanied by enhanced anxiety symptoms (Zimmernan et ai., 2002). There are reports showed that Hypericum also has anxiety activities besides its antidepressant effect. For example, Nicolas' experiments showed that Hypericum was active in preventing the anxiety effect of Mg-depletion animal model (Nicolas et al., 2004). Therefore, in current study, we also tested the potential anxiolytic effects of the hydroalcoholic extracts of these three Hypericum species in established anxiety test model of the elevated plus-maze. As demonstrated in Table 3, both H. hubeiense and H. erectum extracts exerted the anxiolytic activity in the elevated plus-maze similar to that caused by benzodiazepine drug diazepam. However, H. seniavinii did not show significant effect in anxiety improvement. The results indicate the possible involvement of H. hubeiense and H. erectum with benzodiazepine receptor. As the results indicate that the hydroalcoholic extracts of these three Hypericum species have antidepressant activities in mice models with different pattern of activities. H. hubeiense extract at the doses of 250 and 500 mg/kg significantly shortened the immobility time in FST, and significantly shortened the immobility time in TST at the dose of 500 mg! kg. It potentiated the head twitches induced by 5-HT, showed an increase of noradrenaline (NA) toxicity, and also showed anxiolytic activity in the elevated plus-maze test. These activities indicate that the possible mechanisms of action are inhibition of serotonine, and noradrenaline reuptake and binding of GABA receptor. It has the lowest toxicity among the three plants. The multiple mechanism of actions of H. hubeiense is resemble to H. perforatum, which has been shown to shorten the immobility behaviour, inhibit noradrenaline and serotonin re-uptake, inhibit monoamine oxidase, and interact with GABA receptor at a similar dosage (Butterweck et al., 2003).
H. erectum extract significantly shortened the immobility time in FST, and TST. It potentiated the head twitches induced by 5-HT, showed anxiolytic activity in the elevated plus-maze test, but did not increase noradrenaline (NA) toxicity. These activities indicate that the possible mechanisms of action are inhibition of serotonine re-uptake, and binding of benzodiazepine receptor, but not noradrenaline re-uptake. It has relatively high toxicity among the three plants. H. seniavinii extract significantly shortened the immobility time in FST, and TST. It potentiated the head twitches induced by 5-HT, but did not increase noradrenaline (NA) toxicity, or show anxiolytic activity in the elevated plus-maze test. These activities indicate the possible mechanism of action is inhibition of serotonine re-uptake, but not noradrenaline reuptake and binding of benzodiazepine receptor. The dose responses at 250 and 500 mg/kg were not observed for the TST activity of H. erectum extract, and potentiation of head twitches by H.
248
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
hubeiense, while the activities ofthe two extracts in FST and elevated plusmaze test were dose-dependent. These phenomena may be due to the complexicity in chemical composition and pharmacological activities of the herbal extracts. The inverted V-shaped dose-dependent curve has been observed in shortening of immobility by hyperforin (Butterweck et al., 2003). As shown in Table 4, the Hypericum species contain various amounts of hypericin, pseudohypericin and individual flavonoids. The highest content of hypericins and flavonoids (5.76 mg/kg, 40.89 mg/kg respectively) are in H. erectum extract, and the lowest contents are in H. seniavinii extract. Therefore there is no direct correlation of the hypericins content with the antidepressant activity. Further studies are required to define their chemical profiles as well as the relationship between the main constituents and the antidepressant activities. Table 4. Yields ofthe hydroalcoholic extracts ofthree Hypericum species and the contents of the main constituents in the extracts Content in extract
Hypericum
Yield of
species
extract (%)
Hypericins HPLC
Flavonoids IN
H. hubeiense H. seniavinii
14.2 21.8
3.30 1.30
33.76 26.41
H. erectum
15.0
5.76
40.89
(mglg)
In summary the results suggest that these three Hypericum extracts possess antidepressant activity, which may be mainly mediated by serotonergic system. The H. hubeiense and H. erectum zalso possess anxiolytic activity. The results indicate that the three Chinese Hypericum species, in particularly H. hubeiense, possess pharmacological activities similar to H. perforatum and have the potential to be used as new source of antidepressant product.
ACKNOWLEDGEMENTS The authors extend their gratitude to Prof. Chang Hung-Ta and Dr. Wu Lihong for their contribution in identifying the plants, and to Prof. Chen Ruzhu and Prof. Zhu Xiaolan for their support in pharmacological experiments. The study was supported and funded by National Natural Science Foundation of China (2004 No.30470188); and partially supported by the Australia China Special Fund (2007).
REFERENCES Alpermann, H.G., Schacht, u., Usinger, P. and Hock, F.J. (1992). Pharmacological effects of Hoe 249: A new potential antidepressant. Drug Development Research, 25: 267-282.
Antidepressant Activity of the Extracts of Hypericum Species
249
Butterweck, V., Christoffel, V., Nahrstedt, A., Petereit, F., Spengler, B. and Winterhoff, H. (2003). Step by step removal of hyperforin and hypericin: Activity profile of different Hypericum preparations in behavioral models. Life Sciences, 73: 627-639. Fu, P., Zhang, W.D., Liu, RH., Li, T.Z., Shen, Y.H., Li, H.L., Zhang, W. and Chen, H.S. (2006). Two new xanthones from Hypericum Japonicum. Natural Product Research, 20: 1237-1240. Goldstein, D.J., Rampey, A.H., Enas, G.G., Potvin, J.H., Fludzinski, L.A. and Levine, L.R (1994). Fluoxetine: A randomized clinical trial in the treatment of obesity. International Journal of Obesity and Related Metabolic Disorders, 8: 129-135. Goodwin, G.M., Green, A.R and Johnson, P. (1984). 5-HT2 receptor characteristics in frontal cortex and 5-HT2 receptor-mediated head twitch behavior following antidepressant treatment to mice. Brit£sh Journal of Pharmacology, 83: 235-242. Jiang, X., Williams, K.M., Liauw, W.S., Ammit, A.J., Roufogalis, B.D., Duke, C.C., Day, RO. and McLachlan, A.J. (2004). Effect ofSt John's wort and ginseng on the pharmacokinetics and pharmacodynamics of warfarin in healthy subjects. Brit£sh Journal of Clinical Pharmacology, 57: 592-599. Kasper, S. (2001). Hypericum perforatum-A review of clinical studies. Pharmacopsychiatry, 34(Suppl 1): 51-55. Linde, K., Ramirez,G., Mulrow, C.D., Pauls, A., Weidenhammer, W. and Melchart, D. (1996). St John's wort for depression-An overview and meta-analysis of randomized clinical trials. British Medical Journal, 313: 253-258. Lister, R. (1987). The use of a plus-maze to measure anxiety in the mouse. Psychopharmacology, 92: 180-185. McTavish, D. and Heel, RC. (1992). Dexfenfluramine: A review of its pharmacological properties and therapeutic potential in obesity. Drugs, 43: 713-733. Nicolas, S., Catrin, S., Alfred, H., Simone, B.S. and Harald, M. (2004). Magnesium- deficient diet alters depression- and anxiety-related behavior in mice-influence of desipramine and Hypericum perforatum extract. Neuropharmacology, 47: 1189-1197. Porsolt, RD., Le, P.M., Jalfre, M. and Chatterjee, S.S. (1977). Depression: A new animal model sensitive to antidepressant treatments. Nature, 266: 730-732. Sanchez-Mateo, C.C., Bonkanka, C.x., Prado, B. and Rabanal, RM. (2005). Antidepressant properties of some Hypericum canarlense L. and Hypericum glandulosum Ait. extracts in the forced swimming test in mice. Journal of Ethnopharmacology, 97: 541-547. Shank, R.P., Gardocki, J.F., Shcneider, C.R, Vaught, J.L., Settler, P.E., Marvanoff, B.E. and McComsey, D.F. (1987). Preclinical evaluation of McN-5707 as a potential antidepressant. Journal of Pharmacology and Experimental Therapeutics, 242: 74-84. Steru, L.R, Chermat, B. and Thierry, P., Simon. (1985). The tail Suspension Test: A new method for screening antidepressant activity in mice. Psychopharmacology, 85: 367-370. Willner, P. (1984). The validity of animal models of depression. Psychopharmacology, 83: 1-16. Wu, L.H., Hu, H.Y., Huang, S.L. and Yang, D.P. (2002). Review of historical literature records of Forsythia suspensa Vahl and Hypericum perforatum L. China Journal of Chinese Materia Medica, 27: 612-616. Wu, L.H., Yang, D.P. and Wang, F.S. (2004). New taxa of Hypericum (Hypericaceae) in China. Acta Phytotax Sin. 42: 3-8. Zheng, Q.M., Qin, L.P. and Zheng, H.C. (2003), Quantitative analysis of Chemical compositions in 11 Chinese Hypericum plants. Jounal of the Second Military Medicine University, 24: 457-459. Zimmernan, M., Chelminski, I. and McDermut, W. (2002). Major depressive disorder and axis I diagnostic comorbidity. Journal of Climcal Psychiatry, 63: 187-193.
"This page is Intentionally Left Blank"
12 Cytolytic and Toxic Effects of Ostreolysin, a Protein from the Oyster Mushroom (Pleurotus ostreatus) KRISTINA SEPCIC 1 AND ROBERT FRANGEZ 2,*
ABSTRACT
Ostreolysin is a 15 kDa cytolytic protein found in considerable amounts in oyster mushrooms (Pleurotus ostreatus). Ingestion of large quantities of these edible mushrooms can result in adverse reactions, and at least some of these effects can be attributed to ostreolysin. This protein is able to permeabilize erythrocytes and other cells at sub-micromolar concentrations, acting by a colloid-osmotic mechanism and inducing the formation of a membrane pore with a hydrodynamic radius of 2 nm. Its binding to and subsequent permeabilization of the membrane depend on interaction with cholesterolenriched raft-like membrane domains. When injected intravenously into rodents, ostreolysin induces cardiorespiratory arrest and is lethal for mice with an LD50 of 1170 pg/kg. In rats, it induces a transient increase in arterial blood pressure, followed by a progressive drop of blood pressure, associated with noticeable bradicardia and myocardial ischemia. Continued drop of blood pressure is accompanied with ventricular extrasystoles, related to marked hyperkalemia. Moreover, sub-micromolar concentrations of ostreolysin induce a concentration-dependent increase in aortic ring tension. Key words : Cardiorespiratory effect, hemolysis, hyperkalemia, lipid rafts, ostreolysin, Pleurotus ostreatus, toxicity, pore forming protein 1. Department of Biology, Biotechnical Faculty, University of Ljubljana, Vecna pot 111, 1000 Ljubljana, Slovenia. 2. Institute of Physiology, Pharmacology and Toxicology, Veterinary Faculty, University of Ljubljana, Slovenia. * Corresponding author: E-mail: [email protected]
252
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
INTRODUCTION Ostreolysin (Oly) is an acidic 15 kDa protein that is synthesized specifically in the primordia and young fruiting bodies of the oyster mushroom (Pleurotus ostreatus) (Berne et al., 2002). The protein belongs to a larger group of highly homologous proteins called aegerolysins. Aegerolysin-like proteins have been isolated from several mushrooms, moulds, plants and bacteria (Berne et al., 2005). They all have low isoelectric points, similar molecular weights, and are heat-labile. Furthermore, they are stable over a wide range of pH, mainly from 4 to 10 (Sakaguchi et al., 1975; Berne et al., 2005). Several aegerolysins are expressed during the formation of mushroom fruit bodies (Fernandez-Espinar & Labarere, 1997; Berne et al., 2002; Vidic et al., 2005) and during bacterial sporulation (Barloy et al., 1998), which suggests their involvement in the developmental cycle of the organism that produces them. Oly can induce the formation of primordia and development of young fruit bodies of P. ostreatus, even when applied exogenously to the mushroom mycelium (Berne et al., 2007). Binding to natural and artificial lipid membranes followed by permeabilization is another common feature of many aegerolysins. Asphemolysin from the mould Aspergillus fumigatus, as well as mushroomderived aegerolysins (aegerolysin from Agrocybe aegerita, Oly and pleurotolysin A (Ply A) from Pleurotus ostreatus, and eryngeolysin from Pleurotus eryngii) have been reported to lyse erythrocytes and various cell lines (Ebina et al., 1985; Berne et al., 2002; SepCic et al., 2003; Tomita et al., 2004; Ngai & Ng, 2006). The hemolytic and cytolytic effects of aegerolysin-like proteins appear to be associated with their toxicity. One of the most studied aegerolysins, Asp-hemolysin, can be considered as a virulence factor of A. fumigatus during infection of animals by this opportunistic pathogenic mould (Ebina et al., 1982; Malicev et al., 2007). The edible oyster mushroom, although being a massively cultivated species with important medicinal, biotechnological, and environmental applications (Kiies & Liu, 2000; Cohen et al., 2002) has been reported to induce sporadic local intoxications (AlDeen et al., 1987). These toxic effects appear to be connected with Oly, which causes cardiorespiratory arrest after intravenous administration to mice, with a half-lethal dose of 1170 pg/kg CZuzek et al., 2006). The lytic process caused by many pore-forming proteins starts by their recognition of a distinctive membrane component such as a membrane lipid. Aegerolysins show considerable variability in this respect. Asphemolysin, and recombinant PA0122 from Pseudomonas aeruginosa have been suggested to bind specifically to lysophosphatidy1choline (Kudo et al. , 2002; Rao et al., 2008), PlyA to sphingomyelin (SM) (Tomita et al., 2004) and Oly to sense specifically membrane domains enriched together in cholesterol
Effects of Ostreolysin, a Protein from the Oyster Mushroom
253
(Chol) and SM (SepCic et al., 2004). Lateral cell membrane domains, known as lipid rafts, are highly enriched in these two lipids (Pike, 2004). These transient membrane entities, existing in a liquid-ordered (l) state (McConnell & Vrljic, 2003), are more resistant to solubilization by detergents than lipids in liquid-disordered (ld) domains (Lichtenberg et al., 2005) and are therefore often termed detergent-resistant membranes (DRMs) (Simons & Ikonen, 1997). Lipid rafts are involved in several important biological functions, including exocytosis and endocytosis, signal transduction, pathogen entry, and attachment ofligands (Simons and Ikonen, 1997; London, 2002; Edidin, 2003), and stabilize on binding ligand molecules (Kenworthy et al., 2000; Subczynski & Kusumi, 2003; Hancock, 2006). Due to their important functions in the living cell and in cell-cell interactions, there is an increasing need for new techniques to study lipid rafts. Oly, which recognizes specifically the combination of the two main lipid raft components, SM and Chol, is in this regard a good candidate for a new marker of Chol-enriched raft-like membrane microdomains.
MEMBRANE ACTIVITY OF OSTREOLYSIN The ability to bind to natural and artificial lipid membranes and form pores is a common feature of aegerolysin-like proteins. There have been several attempts to express recombinant aegerolysins in bacterial hosts, however most were devoid oflytic potential (Juarez-Perez & Delecluse, 2001; Kumagai et al., 2002; Rao et al., 2008; Rebolj & SepCic, unpublished data). The sole exception is the recombinant form of PlyA, which acts as an active bicomponent cytolysin in concert with the recombinant 59 kDa pleurotolysin B (PlyB) (Sakurai et al., 2004).
Hemolytic and Cytolytic Activities Oly lyses erythrocytes from a variety of species at sub-micromolar concentrations (SepCic et al., 2003). The time course of hemolysis is typical of pore-forming proteins that bind to the membrane in their monomeric form, oligomerize, insert and translocate their polypeptide segments across the lipid bilayers, resulting in the formation of a transmembrane pore consisting of several protein monomers (Gouaux, 1997). The hydrodynamic radius of an Oly-induced transmembrane pore, determined by the use of different osmotic protectants, is 2 nm (SepCic et al., 2003). Some aegerolysins have been shown to form rings and patches at the surface of erythrocyte membranes, detected by scanning electron microscopy (Ebina et al., 1985; Tomita et al., 2004). In the case of pleurotolysin, SDS-stable, ring-shaped 700 kDa complexes were observed when Ply A and Ply B were mixed in a 3:1 molar ratio (Tomita et al., 2004). Incubation of Oly with lipid vesicles formed from total sheep erythrocyte lipids, followed by ultracentrifugation, showed that Oly is located exclusively in the sediment, also pointing to its
254
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
tight binding with lipid membranes. Furthermore, SDS-PAGE analysis of the diluted sediment showed the formation of membrane-bound Oly aggregates of around 34, 64, and 100 kDa, corresponding to dimers, tetramers, and hexamers (SepCic et al., 2003). The pH-optima for Oly binding to the erythrocyte membrane, and for the consequent permeabilization, differ. Optimal binding is usually achieved between pH 6 and 7, while maximal hemolysis is observed between pH 7 and 8 (Berne et al., 2005). Oly however retains its membrane activity over a wide range of pH, from 3.5 to 10.5 (Berne et al., 2005). At physiological pH, the binding of Oly to a membrane correlates with an increase in its intrinsic tryptophan fluorescence and a-helical content (SepCic et al., 2003). Fluorescence titration experiments revealed that Oly changes its conformation more extensively in the lipid-bound than in the free form, suggesting that binding to the bilayers evokes further conformational transitions necessary for membrane insertion and pore formation (Berne et al., 2005). Further, the involvement of intact Oly cysteine residues in the hemolytic process has been proved to be important (Berne et al., 2002, 2005). Lysis can be prevented by micromolar concentrations of some divalent ions, like Hg2+ and Fe 2+ (Berne et al., 2002). Finally, Oly-induced hemolytic effects are abolished by the addition of micromolar concentrations of lysophospholipids (especially lysophosphatidylinositol and sphingosine-I-phosphate), fatty acids (SepCic et al., 2003), and lipid vesicles composed of an equimolar SM:Chol mixture (SepCic et al., 2004). The differing sensitivity of erythrocytes from different species to aegerolysins is another interesting feature (Sakaguchi et al., 1975; Bernheimer & Avigad, 1979; Tomita et al., 2004; Ngai & Ng, 2006). Oly showed no difference in its ability to lyse sheep, bovine and human erythrocytes (SepCic et al., 2003) but was much less potent on those from rodents (Zuiek et al., 2006), dog, and amphibians (Rebolj & SepCic, unpublished data) (Fig 1). In addition to erythrocytes, Oly and several other aegerolysins have been found to be cytotoxic for cell lines at sub-micromolar concentrations (SepCic et al., 2003; SepCic et al., 2004; Malicev et al., 2007; Rebolj et al., 2007) (Table 1). Direct microscopic observation of some tumour cells exposed to Oly showed typical signs of colloid-osmotic lysis, like swelling, blebbing and degranulation of cells (SepCic et al., 2003). Similar events were observed during real-time monitoring of Oly-induced disruption of fluorescently stained chondrocytes and osteoblasts, using confocal microscopy (Malicev et al., 2007). Lytic effects of Oly on different cells can induce hyperkalaemia and are therefore, at least partially, responsible for the observed toxicity of the protein after intravenous administration (Zuiek et al., 2006).
255
Effects of Ostreoiysin, a Protein from the Oyster Mushroom
2.0 1.5 ~
'I c:
I
1.0 0 III
-=: .,....
-o-cow -+-dog -A-rooster -..,-pig - ' 1 - midpuppy -.-frog -·-human -A-sheep -o-rat
/0 0
0.5
o~..----+
0.0 0.1
0.01
Diy w.Mj
Fig 1. Hemolytic activity of ostreolysin on erythrocytes. Hemolysis was assayed by a turbidimetric method (Sepcic et ai., 2003). The reciprocal half-time of hemolysis, (lIt50 ), is plotted against the concentration of ostreolysin, Oly (0.0055 to 3 }lIIl). t50 is the time in which half of the erythrocytes were lysed. Each result is the mean±standard error of three independent experiments Table 1. Cytotoxic properties ofOly on cell lines. ED50 is the concentration of ostreolysin producing 50% cytotoxicity Cell line
ED50 (j1M)
Time of exposure
Reference(s)
HT 1080 (fibrosarcoma, human) MCF 7 (mammalian tumour, human) Chinese hamster ovary cells Human chondrocytes V-79-379A (lung fibroblasts of Chinese hamster) HUVEC (human umbilical vein endothelial cells)
0.67 0.67
2h 2h
SepCic et al., 2003 SepCic et ai., 2003
0.067 0.067 0.087
15 min Ih Ih
0.15
Ih
SepCic et ai., 2004 Malicev et al., 2007 Rebolj et ai., 2007 Rebolj et ai., 2007
Mode of Ostreolysin Binding to Cell Membranes In order to elucidate the structural characteristics of Oly-susceptible membranes, we performed an extensive preliminary study using artificial lipid membranes. Lipid monolayers or bilayers, the latter in the form of vesicles of different sizes, with a range of lipid compositions, were tested for their susceptibility to Oly. Binding was assessed either indirectly in a hemolysis-inhibition assay by incubating Oly with lipid vesicles and subsequent addition of erythrocytes (SepCic et ai., 2003), or directly, using surface plasmon resonance on supported lipid monolayers (SepCic et ai., 2004; Rebolj et ai., 2006; Chowdhury et ai., 2008). Membranepermeabilizing activity was assessed mainly by measuring the leakage of the fluorescent dye calcein from lipid vesicles following the addition of
256
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
Oly (SepCic et al., 2003; Sepcic et al., 2004; Rebolj et al., 2006). We found that lipid vesicles composed of equimolar mixtures of egg phosphatidiylcholine with various other lipids were completely resistant to Oly binding and permeabilization (Sepcic et al., 2003). The only vesicles showing susceptibility to Oly membrane activity were those composed of SM and Chol in a 1: 1 molar ratio (Sepcic et al., 2004). Due to the facts that (i) Oly cannot bind to pure SM or Chol, and (ii) the two lipids are known to be highly enriched in lipid rafts, Oly specific interaction with SM:Chol liposomes suggested its interaction with Chol-enriched raft-like membrane micro domains in living cells. This finding was confirmed by a series of experiments. First, Oly was found in isolated DRMs of both SM:Chol (1:1, mol:mol) vesicles and Chinese Hamster Ovary (CHO) cells (Sepcic et al., 2004). Secondly, Oly bound to and permeabilized SM:Chol membranes in various molar proportions of the two lipids, but only above 30 mol% Chol (Sepcic et al., 2004, Rebolj et al., 2006), the concentration at which this sterol induces the formation of a liquid-ordered phase (de Almeida et al., 2003). Further, binding of sub-lytic concentrations of Oly to various mammalian cells and cell lines (CHO cells, mouse somatotrophs, human chondrocytes and osteoblasts), followed by immunolocalization using fluorescence-labeled antibodies, showed selective binding and clustering of the protein on cell membranes, pointing to its association with distinctive membrane microdomains rich in Chol (Malicev et al., 2007, Chowdhury et al., 2008). Lipid rafts are sensitive to treatment with methyl-~-cyclodextrin (M~CD), which acts as a Chol scavenger and promotes disruption of the structural and functional integrity oflow density membrane domains (Pike & Miller, 1998). In CHO cells pre-treated with 20 mM M~CD (and depleted by 43% of their Chol content), Oly membrane binding was dramatically decreased to only 2%. However, treatment with 10 mM M~CD, which resulted in depletion of membrane Chol by 35%, caused no significant reduction in the binding of Oly (Chowdhury et al., 2008) (Fig 2). In accordance with the results obtained with SM:Chol vesicles in various molar ratios (Sepcic et al., 2004), these data suggest that, to effectively bind and permeabilize lipid membranes, Oly requires a specific threshold membrane Chol concentration. The same trend was observed in experiments with CHO cell mutants having impaired Chol synthesis and consequently a lower fraction of raft-like membrane domains. Binding of Oly to these cells clearly correlated with the amount of their membrane Chol (Sepcic et al., 2004). However, not all our results suggest Oly as a classical lipid raftbinding protein. Our experiments using lipid mono- and bi-Iayers composed of SM and various sterols and Chol derivatives revealed that Oly membrane activity does not completely correlate with the ability of a particular steroid to induce the formation of lo domains (Rebolj et al.,
Effects of Ostreolysin, a Protein from the Oyster Mushroom
120
,
257
100+/-8
100 .!!!.
65+1-4
-----+
80
iii 0
"'C
Q)
c:
~
'0
60 40
#-
20 50+/-7
•
0 0
5
10
15
20
Mf3CD (mM)
Fig 2. Effect of membrane cholesterol concentration on ostreolysin binding to Chinese Hamster Ovary cells. The graph represents percentage of cells able to bind ostreolysin (Oly) following their pre-treatment with methyl-f3-cyclodextrin, Mf3CD, (0 to 20 roM). Symbols represent mean values of Oly bound±standard error from 16-20 micrographs in 2 independent experiments. Numbers adjacent to error bars indicate the percentage of cholesterol in cell membranes. Adapted from Chowdhury et al. 2008
2006). Further, Oly cannot exert its membrane activity on lipid membranes composed of the equimolar mixture of SM, Chol and unsaturated glycerophospholipids that are generally used to mimic the membranes of living cells (SepCic et al., 2004). Finally, immunostaining studies using CHO cells showed that Oly was not co-localized with typical raft-binding proteins like caveolin and cholera toxin B-subunit, suggesting that it binds to distinct Chol-enriched membrane domains (Chowdhury et al., 2008). Altogether, these findings reinforce the emerging idea of the microheterogeneity of the lipid rafts (Roper et al., 2000; de Almeida et al., 2005; Hancock, 2006), comprising at least 2 Chol-enriched domain types - cave olin-containing (caveolae), and caveolin-Iacking rafts (Pike, 2004). It appears that Oly, rather than recognizing the pure lipid component (e.g. Chol or SM), or a specific physical state of the membrane (e.g. a liquid-ordered state), recognizes and binds to specific patterns that are formed by Chol molecules at the surface of biological membranes. This conclusion is reinforced by the fact that the addition of Oly does not affect the electron paramagnetic resonance spectra of equimolar SM:Chol vesicles, which would point to the recruitment of the lipids after the ligand binding (Sepcic et al., 2004).
258
Compo Bio. Nat. Prod., VoL 2 - Efficacy, Safety & Clinical Evaluation I
WHOLE ANIMAL TOXICITY STUDIES OF OSTREOLYSIN AND IN VITRO STUDIES ON TIlE CELL AND ORGAN LEVEL
In vivo Effects of Ostreolysin Despite its putative physiological role during the formation of mushroom fruit bodies (Berne et al., 2002; Vidic et al., 2005; Berne et al., 2007), experiments on erythrocytes and artificial membranes revealed that Oly is also a pore-forming agent (Sepcic et al., 2003). The hemolytic and cytolytic activity of many bioactive proteins from different sources appears to be associated with their toxicity. Many questions arise as to whether these properties are also responsible for Oly toxicity, as demonstrated for another aegerolysin-like protein, Asp-hemolysin (Ebina et al., 1982; Malicev et al., 2007). To investigate this, the toxic and lethal effects of Oly were further studied on experimental rodents in vivo and in vitro. The lethal and toxic effects, expressed as LD50 dose, have been determined in mice and rats, respectively. To determine LD50 , Oly was dissolved in physiological solution and injected as a bolus through the right tail vein of mice. Subsequent clinical signs comprised cyanosis, cessation of movement and hair bristle, and lethal outcome within 20 min for all mice at a dose of 1400 llg/kg or higher CZuiek et al., 2006). The i.v. LD50 in mice was 1170 J.Ig!kg (Zuiek et al., 2006). In order to study its effects on the cardiovascular system and systemic response, in vivo experiments with Oly were performed on adult male Wistar rats. Oly was dissolved in physiological solution and applied as a bolus through the cannulated right jugular vein. Respiratory activity, electrocardiograms (ECG) and blood pressure were monitored. Since Oly is lytic to erythrocytes from various animal species and cell lines (Fig 1), some blood electrolytes were also monitored to find out whether the heart rhythm abnormalities caused by Oly are related to the altered ion concentration (in particular to potassium concentration related to its hemolytic action). In the sub-micromolar range, the toxin lysed rat erythrocytes in vitro in a dose-dependent manner. When injected into rats under general anesthesia, one mice LD50 of Oly causes death in all experimental animals, suggesting that rats are more sensitive than mice. Hemolysis, with a resulting rise in plasma potassium concentration, is probably an important factor in its toxic and lethal effects, as already shown for some other pore forming toxins (Suput et al., 2001; Andrade et al., 2004). In a dose of one mice LD50 , Oly produced a transient pressor response in rats, followed by a prolonged hypotensive response and a progressive, irreversible fall of blood pressure to its mid-circulatory value. In addition, transient apnea is observed after injection of Oly, followed by transient respiratory activity and respiratory arrest (Zuzek, Sepcic & Frangez, unpublished results) (Fig 3). The transient pressor response induced by 1 LD50 of Oly lasted for approximately 1-3 min. The basal mean arterial
Effects of Ostreolysin, a Protein from the Oyster Mushroom
259
blood pressure (MAP) of anaesthetized rats increased from 87 ± 7.6 mm Hg (n=6) before the injection ofOly to 116 ± 9.7 mm Hg (n=6) afterwards. The pressor response was followed by a progressive and irreversible fall in arterial blood pressure (aBP) to the mid-circulatory blood pressure (6-8 mmHg) in all experiments (Fig 3). Oly produced a respiratory arrest in 10-20 s after the toxin injection, followed by a transient respiratory activity after 1-2 min. Two min after injection into anaesthetized rats, Oly significantly reduced the heart rate from 266 ± 14.6 to 121 ± 21.9 (p$;0.05, n=6). At the same time, changes in ECG appeared as sinus arrhythmia, with peaking and increased amplitude of the T-wave, and increased prolonged P-R interval. Elevation ofthe S-T segment, ventricular extrasystoles and widened QRS complexes were also observed regularly, suggesting myocardial ischemia (Fig 3) (Zuzek et al., 2006). Oly binds specifically to raft-like Chol-rich membrane domains (SepCic et al., 2004; Rebolj et al., 2006), followed by the formation of 4 nm transmembrane pores (SepCic et al., 2003). Severe hyperkalemia (10.28 ± 1.25 mmolll, p$;0.05, n=6), caused by the lytic action of Oly on erythrocytes and other cells, may be responsible for bradycardia and irregular heart rate and rhythm in rats (Zuiek et al., 2006). To evaluate the role of cholinergic stimulation of the heart, potentially triggered by the action of Oly on arterial blood pressure and on the ECG, its effects were studied in atropinized rats. Although the effects of Oly on heart rate were less pronounced than in the control group of rats, the muscarinic blocker, atropine, did not significantly abolish or alter the cardiorespiratory effects described for the non-atropinized group of animals injected with Oly. It was therefore concluded that parasympathetic stimulation of the heart plays only a minor role in Oly cardiotoxic action (Zuzek et al., 2006). It was also found that Oly produces the same changes in ECG, blood pressure, and biochemical blood parameters in artificially ventilated rats (n=4), which excludes hypoxia as the primary cause of cardiotoxic effects initiated by respiratory arrest (Zuiek et al., 2006). Although hyperkalemia, suggesting in vivo lysis of erythrocytes and other cells, may be significant for its lethal action, the exact mechanism of the lethal effect of Oly remains unclear. Results from the in vivo experiments (arrhythmias, drop of blood pressure, elevation of S-T segment) suggest myocardial hypoxia as the primary effect. Hence, in order to identify the cellular and molecular mechanisms of the toxic action responsible for in vivo effects of Oly, its effects on vessel ring tension and endothelial cell viability have recently been studied (Rebolj et al., 2007).
Effect of Oly on Isolated Coronary Rings To identify its target organs and cells, in vitro experiments with Oly have been performed on thoracic aortic ring preparations, human umbilical vein
260
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I Oly A~3
H
~
TV
11
aBP
{::o[ ••••••r~'-u.!q) [
I
5 min
I
"t' WI I A
~
ECG
A.
J______l_l"-_J___1_
Fig 3. A representative experiment in which the effects of Oly on electrocardiogram (ECG), arterial blood pressure and breathing were recorded, Tidal volume (TV) and arterial blood pressure (aBP) represent the time course of both parameters recorded during the whole experiment. Al is the ECG recording before the toxin injection and 1\ to A4 the ECG traces after the toxin injection as indicated on the top of the fig
endothelial cells (HUVEC), and lung fibroblasts of Chinese hamster (V-79379A). Oly induced concentration-dependent rat aorta ring contractions at final concentrations of 1 to 50 JJglmL. After the aorta rings were exposed to various concentrations of Oly, endothelium-mediated relaxation of rings pre-contracted with norepinephrine was diminished. This effect may be due to injury of the endothelial cells or to the breakdown of the mechanisms responsible for the acetylcholine (ACh) mediated endothelium relaxation response (Rebolj et al., 2007). This result is in agreement with those related to the lytic effects of O1y on erythrocytes (Sepcic et al., 2003) and umbilical vein endothelial cells (Rebolj et al., 2007). The effective cytotoxic concentration (EC 50 ) of O1y evaluated after 48 h was 1.3 JJglmL for V-79, and 2.2 JJglmL for HUVEC cells in the growth medium (Rebolj et ai., 2007).
Effects of Ostreolysin, a Protein from the Oyster Mushroom
261
The direct damage of endothelial cells probably plays an important role in the cardiotoxicity of Oly CZuzek et al., 2006). This view is additionally supported by the results obtained from the experiments with aortic rings having preserved endothelium, where a functionally defective ACh-mediated relaxation response was clearly demonstrated CRebolj et al., 2007). The final concentrations ofOly (15-30 llg/mL) in the bathing medium, which produced a significant increase in aorta ring tension in vitro, were similar to the maximal calculated concentrations of Oly in the blood during the in vivo experiments. It is suggestive that especially the coronary vessels contraction may be important as the underlying cause of death in experimental rodents (Rebolj et al., 2007). The same effects on coronary vessels could result in a vasospasm leading to ischemia and hypoxic injury of cardiomyocytes. This view is also supported by the ECG data from in vivo experiments CZuzek et al., 2006). Direct harmful effect of Oly on cardiomyocytes and cells of the conductive system of the heart also cannot be excluded.
CONCLUSIONS AND PERSPECTIVES Since Oly may be a causative agent responsible for some documented adverse responses associated with eating oyster mushrooms, it is important to reveal the exact molecular mechanisms responsible for its cardiorespiratory and lethal action observed in rodents. Moreover, although its physiological role in the mushroom is still uncertain, Oly displays a variety of effects that could have valuable applications in various fields of human activity. First, its ability to trigger the fruiting of oyster mushrooms after its application to the mushroom mycelium (Berne et al., 2007) could be used in the field of biotechnology. Secondly, because of its specific interaction with Chol-rich raft-like membrane domains (Septic et al., 2004; Malicev et al., 2007; Chowdhury et al., 2008), Oly could find its place in the field of cell biology as a lipid raft marker. Fluorescence- or spin-labeled raft-binding mutants of cytolytic proteins, devoid of their lytic activity, are good candidates in this regard. One of these (the GM1 ganglioside-binding cholera-toxin B subunit (Bacia et al., 2004) forms part of a commercially available raft-labeling kit. Finally, the cytolytic and hemolytic properties of Oly are comparable to those exerted by Asp-hemolysin, the putative virulence factor of the pathogenic mould Aspergillus fumigatus (Malicev et al., 2007). Studies of the molecular mechanisms of Oly action could therefore shed some light on the action of Asp-hemolysin during the infection, and help in the development of vaccines or in the use of antibodies against infection by A. fumigatus spores, which is very frequent in hospitals associated with surgery. ACKNO~DGEMENTS
The authors gratefully acknowledge Prof. Roger Pain for critical reading of the manuscript, and the Slovenian Research Agency for the financial support.
262
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
REFERENCES Al-Deen, I.H.S., Twaij, H.A.A., Al-Badr, A.A. and Istarabad, T.A.W. (1987). Toxicologic and histopathologic studies of Pleurotus ostreatus mushroom in mice. Journal of Ethnopharmacology, 21: 297-305. Andrade, M.V., Caramez, M.P., Abreu, E.M., Dolnikoff, M., Omar, E.D., Velasco, LT. and Cunha-Melo, J.R. (2004). Lung compliance, plasma electrolyte levels and acid-base balance are affected by scorpion envenomation in anesthetized rats under mechanical ventilation. Comparative btochemistry and physiology. Toxicology & Pharmacology, 138: 97-104. Bacia, K, Scherfeld, D., Kahya, N. and Schwille, P. (2004). Fluorescence correlation spectroscopy relates rafts in model and native membranes. Biophysical Journal, 87: 1034-1043. Barloy, F., Lecadet, M.M. and Delecluse, A. (1998). Cloning and sequencing of three new putative toxin genes from Clostridwm bifermentans. Gene, 211: 293-299. Berne, S., Krizaj, I., Pohleven, F., Turk, T., Macek, P. and SepCic, K (2002). Pleurotus and Agrocybe hemolysins, new proteins hypothetically involved in fungal fruiting. Biochimica et Biophysica Acta, 1570: 153-159. Berne, S., Sepcic, K, Anderluh, G., Turk, T., Macek, P. and Poklar Ulrih, N. (2005). Effect of pH on the pore forming activity and conformational stability of ostreolysin, a lipid raft-binding protein from the edible mushroom Pleurotus ostreatus. Biochemistry, 44: 11137-11147. Berne, S., Pohleven, J., Vidic, I., Rebolj, K, Pohleven, F., Turk, T., Macek, P., Sonnenberg A. and SepCic, K (2007). Ostreolysin enhances fruiting initiation in the oyster mushroom (Pleurotus ostreatus). Mycological Research, 111: 1431-1436. Bernheimer, A.W. and Avigad, L.S. (1979), Cytolytic protein from the edible mushroom, Pleurotus ostreatus. Biochimica et Biophysica Acta, 585: 451-461. Chowdhury, H.H., Rebolj, K, Kreft, M., Zorec, R., Macek, P. and Sepcic, K (2008). Lysophospholipids prevent binding of a cytolytic protein ostreolysin to cholesterolenriched membrane domains. Toxicon, in press. Cohen, R., Persky, L. and Hadar, Y. (2002). Biotechnological applications and potential of wood-degrading mushrooms of the genus Pleurotus. Applied Microbiology and Biotechnology, 58: 582-594. de Almeida, R.F.M., Fedorov, A. and Prieto, M. (2003). Sphingomyelinlphosphatidylcholinel cholesterol phase diagram: boundaries and composition of lipid rafts. Biophysical Journal, 85: 2406-2416. de Almeida, R.F., Loura, L.M., Fedorov, A. and Prieto, M. (2005). Lipid rafts have different sizes depending on membrane composition: a time-resolved fluorescence resonance energy transfer study. Journal of Molecular Biology, 346: 1109-1120. Ebina, K, Yokota, K and Sakaguchi, O. (1982). Studies on the toxin of Aspergillus fumigatus XIV. Relationship between Asp-hemolysin and experimental infection of mice. Japanese Journal of Medical Mycology, 23: 246-252. Ebina, K, Ichinowatari, S. and Yokota, K. (1985). Studies on toxin of Aspergillus fumigatus. XXII. Fashion of binding of Asp-hemolysin to human erythrocytes and Asp-hemolysinbinding proteins of erythrocyte membranes. Microbiology and Immunology, 29: 91-101. Edidin, M. (2003). The state of lipid rafts: from model membranes to cells. Annual Review of Btophysics and Btomolecular Structure, 32: 257-283. Fernandez Espinar, M.T. and Labarere, J. (1997). Cloning and sequencing of the Aa-Pril gene specifically expressed during fruiting initiation in the edible mushroom Agrocybe aegerita, and analysis of the predicted amino-acid sequence. Current Genetics, 32: 420-424. Gouaux, E. (1997). Channel-forming toxins: tales of transformations. Current Opinion in Structural Biology, 7: 566-573. Hancock, J.F. (2006). Lipid rafts: contentious only from simplistic standpoints."Nature Reviews. Molecular Cell Btology, 7: 456-462. Juarez-Perez, V. and Delecluse, A. (2001). The Cry toxins and the putative hemolysins of Clostridium bifermentans ser. malaysia are not involved in mosquitocidal activity. Journal of Invertebrate Pathology, 78: 57-58.
Effects of Ostreoiysin, a Protein from the Oyster Mushroom
263
Kenworthy, A.K., Petranova, N. and Edidin, M. (2000). High-resolution FRET microscopy of cholera toxin B-subunit and GPI-anchored proteins in cell plasma membranes. Molecular Biology of the Cell, 11: 1645-1655. Kiies, D. and Liu, Y. (2000). Fruiting body production in basidiomycetes. Applied Microbwlogy and Biotechnology, 54: 141-152. Kudo, y., Ootani, T., Kumagai, T., Fukuchi, Y. and Ebina, K (2002). A novel oxidized lowdensity lipoprotein binding protein, Asp-hemolysin, recognizes lysophosphatidylcholine. Biological & Pharmaceut£cal Bulletin, 25: 787-790. Kumagai, T., Kudo, Y., Fukuchi, Y., Ebina, K. and Yokota, K. (2002). Expression of a synthetic gene encoding the Asp-hemolysin from Aspergillus fum£gatus in Eschenchia coli. Biological & Pharmaceutical Bulletin, 25: 115-7. Lichtenberg, D., Goni, F.M. and Heerklotz, H. (2005). Detergent-resistant membranes should not be identified with membrane rafts. Trends in Biochem£cal Sciences, 30: 430-436. London, E. (2002). Insights into lipid raft structure and formation from experiments in model membranes. Current Opinion in Structural Biology, 12: 480-486. Malicev, E., Chowdhury, H., Macek, P. and SepCic, K. (2007). Effect of ostreolysin, an Asp-hemolysin isoform, on human chondrocytes and osteoblasts, and possible role of Asp-hemolysin in pathogenesis. Medical Mycology, 45: 123-130. McConnell, H.M. and Vrljic, M. (2003). Liquid-liquid immiscibility in membranes. Annual Review of Biophysics & Biomolecular Structure, 32: 469-492. Ngai, P.H.K. and Ng, T.B. (2006). A hemolysin from the mushroom Pleurotus eryngii. Apphed M£crobiology and Biotechnology, 72: 1185-1191. Pike, L.J. (2004). Lipid rafts: heterogeneity on the high seas. Bwchem£cal Journal, 378: 281-292. Pike, L.J. and Miller, J.M. (1998). Cholesterol depletion delocalizes phosphatidylinositol bisphosphate and inhibits hormone-stimulated phosphatidylinositol turnover. Journal of Biolog£cal Chemistry, 273: 22298-22304. Rao, J., DiGiandomenico, A., Dnger, J., Bao, Y., Polanowska-Grabowska, R.K. and Goldberg, J.B. (2008). A novel oxidized low-density lipoprotein-binding protein from Pseudomonas aeruginosa. Microbiology, 154: 654-665. Rebolj, K., Poklar Dlrih, N., Macek, P. and Sepcic, K. (2006). Steroid structural requirements for interaction of ostreolysin, a lipid-raft binding cytolysin, with lipid monolayers and bilayers. Biochimica et Bwphysica Acta, 1758: 1662-1670. Rebolj, K., Batista, D., SepCic, K., Cestnik, V., Macek, P. and Frangez, R. (2007). Ostreolysin affects rat aorta ring tension and endothelial cell viability in vitro. Toxicon, 49: 1211-1213. Roper, K., Corbeil, D. and Huttner, W.B. (2000). Retention of prominin in microvilli reveals distinct cholesterol-based lipid micro-domains in the apical plasma membrane. Nature Cell Biology, 2: 582-592. Sakaguchi, 0., Shimada, H. and Yokota, K. (1975). Purification and characteristics of hemolytic toxin from Aspergillus fumigatus. Japanese Journal of Medical Sciences and Biology, 28: 328-331. Sakurai, N., Kaneko, J., Kamio, Y. and Tomita, T. (2004). Cloning, expression, and poreforming properties of mature and precursor forms of pleurotolysin, a sphingomyelinspecific two-component cytolysin from the edible mushroom Pleurotus ostreatus. Bwchimica et Biophysica Acta, 1679: 65-73. SepCic, K., Berne, S., Potrich, C., Turk, T., Macek, P. and Menestrina, G. (2003). Interaction of ostreolysin, a cytolytic protein from the edible mushroom Pleurotus ostreatus, with lipid membranes and modulation by lysophospholipids. European Journal of Bwchemistry, 270: 1199-1210. SepCic, K., Berne, S., Rebolj, K., Batista, D., Plemenitas, A., Sentjurc, M. and Macek, P. (2004). Ostreolysin, a pore-forming protein from the oyster mushroom, interacts specifically with membrane cholesterol-rich lipid domains. FEBS Letters, 575: 81-85. Simons, K and Ikonen, E. (1997). Functional rafts in cell membranes. Nature, 387: 569-572. Subczynski, W.K. and Kusumi, A. (2003). Dynamics of raft molecules in the cell and artificial membranes: approaches by pulse EPR spin labeling and single molecule optical microscopy. Bwchimica et Biophystca Acta, 1610: 231-243.
264
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Suput, D., Frangez, R. and Bunc, M. (2001). Cardiovascular effects of equinatoxin III from the sea anemone Actmia equina (L.). Toxicon, 39: 1421-1427. Tomita, T., Noguchi, K., Mimuro, H., Ukaji, F., Ito, K., Sugawara-Tomita, N. and Hashimoto, Y. (2004). Pleurotolysin, a novel sphingomyelin-specific two-component cytolysin from the edible mushroom Pleurotus ostreatus, assembles into a transmembrane pore complex. Journal of Biological Chemistry, 279: 26975-26982. Vidic, I., Berne, S., Drobne, D., Macek, P., Frangez, R., Turk, T., Strus, J. and SepCic, K. (2005). Temporal and spatial expression of ostreolysin during development of the oyster mushroom (Pleurotus ostreatus). Mycological Research, 109: 377-382. Zuzek, M.C., Macek, P., SepCic, K., Cestnik, V. and Frangez, R. (2006). Toxic and lethal effects of ostreolysin, a cytolytic protein from edible oyster mushroom (Pleurotus ostreatus), in rodents. Toxicon, 48: 264-271.
13 Antioxidant, Antisecretory and Gastroprotective Activities from Leiothrix flavescens THIAGO DE MELLO MORAES 1, MARCELO APARECIDO SILVA2, CLENILSON MARTINS RODRIGUES 2, LOURDES CAMPANER DOS SANTOS 2, MIRIAM SANNOMIYA2 , LUCIA REGINA MAcHADO DA ROCHA 1, ALBA REGINA MONTEIRO SOUZA BRIT03 , TAIS M. BAUAB 4 , WAGNER VILEGAS 2 AND CLl!;LIAAKIKO HIRVMA-LIMAl,*
ABSTRACT Leiothrix flavescens, popularly know as "sempre viva" belongs to Eriocaulaceae family with important biological activity. Extracts of this specie contain mainly flavones and xanthones. The methanolic extract (MeOH) obtained the scapes of L. flavescens was evaluated for its ability to protect the gastric mucosa against gastric mucosa injuries agents in rodents such as HClIEtOH, non-steroidal anti-inflammatory drug or stress. We also evaluated its activity against Helicobacter pylori and its antioxidant property. All pretreatment with MeOH extract from L. flavescens show significant gastroprotective action. No acute toxicity was observed in animals treated with MeOH extract at doses of 5 g/kg (p.o.). The MeOH extract from L. flavescens also increased intestinal motility and changed gastric juice parameters significantly by decreasing gastric acid secretion and promoting reduced acid output. The MeOH extract also shown effective action against H. pylori by inhibiting their growth and presented an interesting antioxidant action in vitro. The 1. Physiology Department, Biosciences Institute, Sao Paulo State University-UNESP, c.p. 510; Zip Code: 18618-000, UNESP, Botucatu, SP, Brazil. 2. Organic Chemistry Department, Chemistry Institute, Sao Paulo State UniversityUNESP, c.p. 355; Zip Code: 14800-900, UNESP, Araraquara, SP, Brazil. 3. Physiology and Biophysics Department, Biology Institute-UNICAMP, c.p. 6109; Zip Code: 13083-970, UNICAMP, Campinas, SP, Brazil. 4. Biological Sciences Department, Sao Paulo State University-UNESP, c.p.355; Zip Code: 14800-900, UNESP, Araraquara, SP, Brazil. * Corresponding author: E-mail: [email protected].
266
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
present data show the effectiveness of MeOH extract from L. fiavescens as antacid/antiulcer and laxative. Key words: Flavonoids, antiulcer, Leiothrix fiavescens, xanthones, antiHelicobacter pylori, antioxidant action
INTRODUCTION Leiothrix Ruhland is an exclusively South American genus of about 37 species mainly restricted to Brazil, with only two species occurring elsewhere: Leiothrix fiavescens (Bong.) Ruhland that occurs in Brazil, Venezuela, Guyana and Peru, and Leiothrix celiae Moldenke which is found only in Venezuela (Giulietti et al., 1998). The other species are mostly endemic to small areas in Minas Gerais and in Bahia, states of central Brazil (Giulietti et al., 1994). This genus and other species of the Eriocaulaceae family such as Syngonanthus and Paepalanthus are known as "sempre vivas". Chemical studies on species of Leiothrix are also scarce. Salatino et al. (1990) studied the contents of soluble phenolic compounds of the capitula of 21 species of Leiothrix. The presence of nepetin-7 -O-P-Dglucopyranoside, nepetin-7 -O-P-D-arabinopyranoside and luteolin 0- and C-glucopyranoside were determined in Leiothrix species (Dokkedal & Salatino, 1992). Another study determined the presence of xanthones and flavonoids with antioxidant activity in capitula of Leiothrix species (Santos et al., 2001). Although there has been no pharmacological study about this species, the Eriocaulaceae family has presented significant effects on gastrointestinal tract. The antiulcerogenic activity of two species of Syngonanthus (Coelho et al., 2006; Batista et al., 2004) has also been determined. Di Stasi et al. (2004) reported the intestinal anti-inflammatory activity of paepalantin, an isocoumarin isolated from Paepalanthus bromelioides (Eriocaulaceae).Therefore, the present work was carried out in order to investigate the gastric effect of the methanolic extract (MeOH) from scapes of L. fiavescens against acute experimental models of gastric ulcer in rodents as well as to perform its phytochemical investigation.
MATERIALS AND METHODS Plant Material Scapes of Leiothrix fiavescens (Bong.) Ruhland were collected in May 1997, at Serra do Cipo, Minas Gerais state, Brazil and authenticated by Professor Dr. Paulo Takeo Sano from Instituto de Biociencias (IB) da Universidade de Sao Paulo (USP), Sao Paulo. A voucher specimen (CFCR n° 4680) was deposited at the Herbarium of the IB-USP.
Antioxidant Activities from Leiothrix flavescens
267
Animals Male Swiss albino mice (25-35 g) from the Central Animal House of the Universidade Estadual Julio de Mesquita Filho (UNESPlBotucatu) were used. The animals were fed a certified Nuvilab CR-a® (Nuvital) diet with free access to tap water under standard conditions of 12 h dark-12 h light and temperature (22 ± 1%). Fasting was used prior to all assays because standard drugs and extract were always administered either orally (by gavage) or by intraduodenal route. The protocols followed the recommendations of the Canadian Council on Animal Care (Olfert et al., 1993).
Drugs and Chemicals Hydrochloric acid (Nuclear, Brazil), cimetidine, atropine (Sigma Chemical Co., St Louis, MO, USA), piroxicam (Hexal, Brazil) and lansoprazole (Medley, Brazil) were used in this study. Extract was dissolved in NaCI solution 0.9% (vehicle). All substances were prepared immediately before use and the reagents used were of analytical grade.
Acute Toxicity The acute toxicity studies of L. flavescens were performed in mice. In this assay, increasing doses ofMeOH extract were orally administered to groups of ten animals for each dose after a 6 h fast. Animals receiving the vehicle served as control. The signs and symptoms associated with the MeOH administration were observed at 0, 30, 60, 120, 180 and 240 min. after and then once a day for the next 14 days. At the end of the period the number of survivors was recorded. The acute toxicological effect was also estimated (Souza Brito, 1995).
Antiulcerogenic Effect HClIethanol-induced ulcer: The antiulcerogenic activity obtained from L. flavescens was studied in HClIethanol-induced gastric ulcer (Mizui & Doteuchi, 1983). Mice were divided into groups of 7-8 animals, which fasted 24 h prior to receiving an oral dose of the vehicle, saline (10 mU kg), lansoprazole (30 mg/kg) or L. flavescens (250, 500 or 1000 mg/kg). After 50 min. all groups were orally treated with 0.2 mL of a 0.3 M HClI 60% ethanol solution (HClIethanol) for gastric-ulcer induction. Animals were killed 1 h after the administration of HClIethanol, and the stomachs excised and inflated by saline injection (2 mL). The extent of the lesions was measured using ulcerative lesion index (ULl) and pH of gastric juice determined. This index was expressed as the sum of all lesions (Szelenyi & Thiemer, 1978).
268
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
Non-Steroidal Antiinflammatory Drug (NSAlD)-Induced Gastric mcers In this model (Puscas et al., 1997), gastric ulcer was induced using piroxicam (30 mg/kg, s.c.) administered to mice. The MeOH extract from L. flavescens (250, 500 or 1000 mg/kg), cimetidine (100 mg/kg) or saline were administered orally 30 min before the induction of gastric ulcer. The animals were killed 4 h after treatment with the ulcerogenic agent and gastric damage was determined as described above.
Hypothermic Restraint Stress mcer Mter fasting, the mice received an oral administration of MeOH extract from L. flavescens (250, 500 or 1000 mg/kg) , cimetidine (100 mg/kg) or saline (10 mUkg). 1 h after treatment, mice were immobilized in a restraint cage at 4°C for 4 h to induce gastric ulcer. The animals were killed and gastric lesions were determined as described above (Levine, 1971).
Determination of Gastric Secretion All groups of mice fasted for 24 h, with free access to water. Immediately after pylorus ligature, MeOH extract from L. flavescens (250, 500 or 1000 mg/kg) , cimetidine (100 mg/kg) as positive control, or the vehicle was administered intraduodenally. The animals were killed 4 h later and the abdomen was opened and another ligature placed around the esophagus close to the diaphragm. The stomachs were removed and the gastric content collected to determine the pH values and total amount of gastric-juice acid. Distilled water was added and the resultant solution centrifuged at 2500 G for 10 min. Total acid in the gastric secretion was determined in the supernatant volume by titration to pH 7.0 with 0.01 N NaOH (Shay et al.,1945).
Anti-Helicobacter pylori Activity The MeOH of L. flavescens was tested to detect anti-Helicobacter pylori activity (Hachem et al., 1996). The stain of Helicobacter pylori (ATCC 43504) was isolated from patients with duodenal ulcer disease. Frozen H. pylori isolate was thawed and grown on 5% sheep blood agar plates for 3 to 4 days at 37°C in 10% CO 2 and 98% humidity. Each plate was swabbed with a sterile cotton-tipped applicator and the cells were suspended in sterile saline to obtain turbidity equivalent to a 2.0 McFarland standard. MullerHinton broth containing 10% horse serum was added to all wells of a 96well microtiter plate (Corning-USA). Each well was incubated with H. pylori at a final concentration of 1 x 105 CFU/mL. The plates were incubated for 5 days in a microaerobic atmosphere at 37°C. Following incubation, the plates were examined visually and spectrophotometrically and the lowest concentration showing complete inhibition of growth was recorded as the MIC (minimum inhibitory concentrations). Staphylococcus aureus ATCC
Antioxidant Activities from Leiothrix flavescens
269
25923 and E.coli ATCC 25922 were used as control organisms for clarithromycin and ampicillin, respectively. The results were considered valid only when the MIC values for the control organisms were within the ranges established by the National Committee for Clinical Laboratory Standards (NCCLS).
Determination ofActivity on Intestinal Motility Mice submitted to a 6 h fast were treated orally with the vehicle (saline, p.o.), atropine (1 mg/kg, p.o.), MeOH (250, 500 or 1000 mg/kg, p.o.). 30 min after the treatments, all animals received 10% activated charcoal (0.1 mU 10 g, p.o.) and were killed 30 min later. The distance traveled by activated charcoal up to the last portion of small intestine was determined (Stickney & Northup, 1959). The results were expressed as percentage of the total length of the small intestine traveled by this marker.
Antioxidant Activity The antioxidant activity was evaluated through the methodology of lipidic peroxidation with membrane of brain from Wi star rats (Stocks, 1974), with a serial dilution of the methanolic extract of L. fiavescens (1,14; 2, 27; 4, 55; 9, 09; 18, 18 e 36, 36 )lg/mL) with the purpose to obtain IC 50 (inhibitory concentration of 50% of the oxidation) through the production of malonidialdehide (MDA) (Fee & Teitelbaum, 1972).
Statistical Analysis Results were expressed as the mean ± S.E.M. Statistical significance was determined by one-way analysis of variance followed by Dunnett's test, with the level of significance set at p<0.05.
Extraction and Isolation The scapes ofL. flavescens (500 g) were separated from the capitula, powdered and successively extracted with chloroform, methanol and 80% methanol (1 week for each solvent). The solvents were evaporated in a vacuum yielding black syrups. A portion (2.0 g) of the ethanol extract of L. flavescens was submitted to CC on sephadex LH-20 (100 x 5 cm), with MeOH as eluant. Fractions (8 mL) were collected and checked by TLC on silica gel in Si gel plates, n-BuOH-AcOH-H20 (20:5:8, v/v/v)]. Fractions 90-100 (40 mg) were purified by repeated CC on polyvinylpolypyrrolidone (Sigma) eluted with MeOH yielding compounds 1 (7.0 mg) and 2 (9.0 mg). Fractions 65-73 (69.8 mg) were further purified by HPLC using MeOH:H 20 (1:1, v/v) as eluant to afford 3 (4.2 mg, RT = 8 min), and 4 (10.8 mg, RT = 11 min). Fractions 47-51 (12.0 mg) using MeOH:H 20 (2:3, v/v) as eluant gave compound 5 (6.5 mg, RT = 18 min). Fractions 40-46 using MeOH:H20 (2:3, v/v) as eluant afforded compound 6 (8.5 mg, RT = 9 min) and 7 (5.8 mg, ~ = 16 min.). Fractions 8283 contained pure 8 (13.3 mg). Substances (Fig 1) were identified by NMR (nuclear magnetic resonance, as 13C-NMR, IH-NMR), spectra and by
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
270
comparison with previous data reported in the literature (Santos et al., 2001; Agrawal, 1989; Harborne, 1996).
OR
OR 1,2,4-8 Compounds
Reference(s)
Name
Rl
R2
R3
1 2 4
H H OH
H H glc
OH H OH
5
glc
CH3
OH
6
CH3
glc
OCH 3
7
glc
H
H
8
OH
H
OH
(Agrawal, Luteolin Apigenin 1989; Harborne, 3',4' ,5,6-tetrahydroxy-71996) methoxy-flavone 3',4',5-triihydroxy-7-metoxy6-C-glucopyranosylflavone 4',5-dihydroxy-3',6-dimethoxy7-O-~-D _glucopyranosylflavone 3',4',5, 7-tetrahydroxy-6-Cglucopyranosylflavone 3',4',5,6,7,8-hexahydroxyflavone
COOCH 3 0
HO
OH
OH OH
3- 8-carboxy-methyl-l,3,5,6-tetrahydroxyxanthone (Santos et al., 2001). Fig 1. Flavonoids and xanthones isolated from the scapes of Leiothrix flavescens
Experimental Section NMR spectra in DMSO-d 6 were obtained using a Varian INOVA 500 spectrometer operating at 500 MHz for IH and 150 MHz for 13C. HPLC separations were achieved on a Chance HPLC System equipped with a R401 refractive index detector and with a phenomenex Luna reversed phase C-18 columns (250 x 10 mm x 10 mm) and Rheodyne injector. TLC were performed on silica gel SiF254 (Merck) eluted with CHCI 3:MeOH:H2 0 (80:18:2, v/v/v) and n-BuOH:HAc:H 2 0 (65:35:25, v/v/v), the plates were visualized using UV light (254 and 365 nm).
Antioxidant Activities from Leiothrix flavescens
271
HPLC-UV-PDA Analysis The chromatography profile of the methanolic extract of L. flavescens was established with a liquid chromatograph equipped with a PU-2089 quaternary solvent pump, a MD-2010 photo diode array detector (Jasco, Tokyo, Japan) and a Rheodyne 7725 sample injector with a 20 1I1 sample loop (Rheodyne, Cotati, CA, USA). The analytical column was a Phenomenex Luna RP18 (250 x 4.6 mm lD. x 5 lIm, equipped with a Phenomenex security guard 4.0 x 2.0 mm, Torrance, CA, USA). The mobile phase composition used was: water (eluent A) and acetonitrile (eluent B), both with 0.05% ofTFA. The gradient program was as follows: 15-22% B (25 min.), 22-40% B (15 min.), 40-78% B (5 min.), 78-100% B (5 min.) and 100% B isocratic (5 min.). Total run time was 55 min. The flow rate of the mobile phase was 1.0 mUmin. EZChrom Elite Data System software was used for both the operation of detector and for data processing. The methanolic extract (2 mg) was dissolved in 2 mL methanol, filtered through a 0.45 11m membrane PTFE filter (Millex), resuspended in 3 mL of water and 20 I!l submitted to the analysis by HPLC. The HPLC profile of the methanolic extract prepared from scape of L. flavescens was presented in Fig 2. r---
jA
500
1
,]j E
!
Minutes
r~~'~~~\?'\J ,~!f/~\ .. j "'Ie
:lOt ____ ~~______ 2tO
220
a... :no
2M
)DO
m
:MIl
HO
310....
,..10
,I ___ ~_~___ 2DI
no
240
HI
280
_
321
,...
3M
310....
"E ~ 0 __ ~ __. ___ ~ _ no
Z20
241
2M
210
)DO
m
)40
310
* ....
Fig 2. (A) HPLC fingerprint profile of crude methanolic extract from scape ofL. flavescens. Retention times of major peaks are indicated in chromatogram. B) The regions of xanthones are shown as insets. Representative UV for the classes of compounds found in the methanolic extract is shown in the lower part ofthe chromatogram. C) Flavones derivatives, D) Cafeoyl quinic derivatives and E) xanthones derivatives
272
Camp. Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
RESULTS AND DISCUSSION Biological experiments may be designed with the objective of determining the dose of a chemical required to produce any specific effect. The measured effect need not be death of the animal but may be any type of biologic effect that can be quantified. So as part of the pharmacological evaluation of MeOH extract of L. flavescens, we investigated the acute toxicity in mice. A single oral administration of MeOH extract (5,000 mglkg) did not produce any sign or symptom of toxicity in the treated animals. After 14 days of extract administration, no animal died and no significant changes in daily body or organ weight were observed. There were no significant alterations in water or food consumption. In autopsy, no significant change or lesion was observed in the viscera of each animal. Loomis & Hayes, (1996) described the classification of some chemicals agents with categories of toxicity and chemicals may be categorization by practically nontoxic at dose of 5 glkg. Thus, this result indicates that the MeOH extract from E. ligulatum has no acute toxicological effect when administered orally. These results guarantee the continuance from the pharmacological studies from this specie. For this reason, a 5-fold lower dose (1000 mglkg) was used as the maximum doses in all experiment to determine the general profile of antiulcer effect of extract from L. flavescens scapes. We evaluated the preventive effect of three oral doses of 250, 500 or 1000 mglkg of the MeOH extract obtained from L. flavescens administered to mice using different standard experimental models in order to verify their action on the gastrointestinal tract. This approach allows a general profile to be established for the gastroprotective activity from the vegetal extract for a future investigation of its action mechanism. In the HClIEtOH-induced gastric ulcers, the lesions were characterized by multiple-hemorrhage red bands of different sizes along the long axis of the glandular stomach. As shown in Fig 3, all doses of the MeOH extract showed significant antiulcer activity. The above results observed with MeOH extract (250, 500 or 1000 mg/kg) demonstrated significant inhibition of ulcerative lesion by 51% (24 ± 4.1 mm), 76% (12 ± 4.1 mm) and 87% (6.2 ± 1.9 mm), respectively, compared to the control value (49 ± 7.7 mm). The ability of the gastric mucosa to resist to injury caused by endogenous secretions (acid, pepsin and bile) and by exogenous irritants (e.g. alcohol) can be attributed to a number of factors that have been collectively referred to as mucosal defense (Kinoshita et al., 1995). The formation of gastric mucosal lesions by necrotizing agents such as HCl and EtOH has been reported to involve the depression of these gastric defensive mechanisms and also promote stasis in gastric blood flow that contributes to the development of the hemorrhagic and necrotic aspects of tissue injury (Morimoto et al., 1991).
60
• vehicle D positive control
50
~
40
MeOH 250 mg/kg
I'TI MeOH 500mg/kg 30
lID MeOH 1000 mg/kg
20 10 0 HCl/ethanol
piroxicam
Stress
Fig 3. Effects of oral treatments with MeOH extract from L. fZavescens on ulcerative lesion index (ULI) in gastric lesions induced by HClIethanol, NSAID (piroxicam) and hypothermic-restraint-stress in mice Data are presented as mean ± S.E.M. ANOVA: F(4,32) = 82.8 (p<0.05) for pH-HClIethanol; F(4,32) = 8.88 (p<0.05) for ULI-HClIethanol; F(4,44) = 4.85 for pH-NSAIDS (p<0.05); F I4,44) = 6.27 for ULI-NSAIDS (p<0.05); F I4 ,25) = 0.74 (p>0.05) for pH-stress model and F(4,25) = 6.45 (p<0.05) for ULI-stress model. Dunnett's test: * p<0.05, **p
t>:> -.1
<:.:>
274
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Gastric lesions induced by ethanol are not inhibited by antisecretory agents such as cimetidine, but are inhibited by agents that enhance mucosal defensive factors such as prostaglandins (Garner, 1992). These results shown that the MeOH extract of L. flavescens probably has an antiulcerogenic effect related to the cytoprotective action since this extract protected against the necrotizing action of ethanol. Nonsteroidal anti-inflammatory drugs (NSAIDs) such as aspirin®, piroxicam and indomethacin remain among the most commonly used pharmacological agents (Garner, 1992). However, these classes of substances cause gastrointestinal ulceration. This effect is correlated to the ability of these agents to suppress the prostaglandin synthesis (Wallace, 2001). Fig 3, shows the antiulcerogenic effect ofMeOH extract in the NSAIDinduced lesion model. Significant reductions in the damage to gastric mucosa (p<0.05) of 41% (250 mglkg) , 57% (500 mglkg) and 72% (1000 mg! kg), respectively, were observed with the use of the MeOH extract when compared to the control value. The increased synthesis of mucous and/or prostaglandins can be the probable cytoprotective mechanism involved with this activity (Guth et al., 1984). Stress is the major causes of gastric disturbances in modern society. Gastric stress ulceration is probably mediated by histamine release with enhanced acid secretion and reduced mucous production. Moreover, stressinduced ulcer can be prevented partially or entirely by vagotomy. Vagal over-activity has been suggested to be the principal factor in stress-induced ulceration (Singh & Majumdar, 1999). In the gastric ulcer induced by hypothermic-restraint stress (Fig 3), the MeOH extract also showed significant gastroprotection (p<0.05); and the gastric-lesion inhibition for the extract were 60% (500 mglkg) and 70% (1000 mglkg) respectively, when compared with the vehicle. Ares et al. (1995) identified gastroprotective properties in the flavone class and phytochemical analyses of L. flavescens shown that this species produces mainly flavones that certain contributed to gastroprotective action of this plant (Silva et al., 2007). In the next step, we investigated the biochemical alterations promoted by L. flavescens extract in gastric-juice parameters. The MeOH extract was administered intraduodenally after submitting the mice to pylorus ligature (Table 1). Ligation of the pylorus for 4 h produced accumulation of gastric juice and lesion in gastric mucosa, whereas the intraduodenal route showed systemic action for all of the treatments. As shown in Table 1, the MeOH extract significantly decreased the gastricacid secretion (p<0.05) and promoted reduced acid output (p
Antioxidant Activities from Leiothrix flavescens
275
arthrotrichus were observed only by flavonoids rich fraction. This results obtained with MeOH extract of L. flavescens as anti secretory agents is important as prophylaxis of ulcer gastric and gastric carcinogenesis. But gastric carcinogenesis is a complex, multistep and multifactorial event, in which the role of Helicobacter pylori infection has been established. We also evaluated the anti-Helicobacter pylori action of MeOH extract of L. flavescens. In the course ofthe study the MeOH extract was found to present an important antibacterial activity against the standard strain (ATCC 43504). The results showed that MIC of MeOH against H.pylori was 125 Ilg/mL. The literature reported that MIC<500 Ilg/mL is considered interesting for vegetal extracts (Gadhi et al., 2001; Hachem et al., 1996). We conclude that the MeOH has an excellent antimicrobial action against one of the most important factors that cause gastric ulceration. Table 1. Effects of MeOH extract from L. flavescens administered intraduodenally (i.d.) on the biochemical parameters of gastric juice obtained from pylorusligature mice Treatment group (p.o.) Control Cimetidine L. flavescens
Dose (mg/kg)
100 250 500 1000
N
9 5 6 6 6
pH (unit) 2.78 3.75 4.00 3.67 3.50
± ± ± ± ±
0.40 0.75 0.45 0.76 0.62
Gastric juice volume (mU4h) 1.55 1.12 1.08 0.94 0.72
0.13 0.11 0.18 0.17 * ± 0.11 ** ± ± ± ±
Acid output (mEqlmU4h) 25.8 15.7 18.3 3.25 5.00
± ± ± ± ±
2.29 3.71 * 2.33 0.48 ** 0.91 **
The results are the mean±S.E.M. N: number of mice used. ANOVA, F(4.27) = 0.80 (p>0.05) for pH; F(4.27) = 5.17 for gastric juice volume (p>0.05); F(4.27) = 18.4 (p<0.05) for [H+]. Dunnett's Test: * p<0.05; ** p
Farinati et al. (2008) shown that among the pathways relevant to gastric carcinogenesis and correlated with H. pylori infection, the production of reactive oxygen species may be quite important. The eradication of H. pylori to prevent the production of reactive oxygen species and accumulation of oxidative DNA damage are strategic point to prevent gastric ulcer and gastric carcinogenesis. The reactive oxygen species (ROS) generate by metabolism of arachidonic acid, platelets, macrophages and smooth muscle cells contribute to gastric mucosal damage (Repetto & Llesuy, 2002). MeOH extract from L. flavescens has an antioxidant activity in vitro with IC 50 of 21.55 ± 0.02 Ilg/mL. This value is considered a good antioxidant activity when it comes from a crude extract and thus justifying the anti ulcerogenic activity observed in other models. We also observed that MeOH extract from L. flavescens was able to increase the propulsion of charcoal meal in a dose dependent manner (Table 2). The velocity of intestinal travel is one of the factors that affected the intensity of luminal absorption and regulated the biodisponsibility of the
276
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
drug administered orally. Thus gut transit test with activated charcoal was carried out to determine the effect of MeOH from L. flavescens on intestinal motility. This result indicated that MeOH extract from L. flavescens also has a laxative property. There are few laxative products related to intestinal peristalsis that have been in use for decades. Many of these products in use today are of plant origin (Fintelmann, 1991). Although laxative effects have been described for some antiulcer drugs (Motiva et al., 1996; Bobrove, 1990), such effects of MeOH extract is being currently investigated by our group. Table 2. Effects of MeOH extract from L. flavescens on intestinal transport, induced by charcoal, in mice (n=71 Treatment group (p.o.) Control Atropine L. flavescens
Dose (mglkg) 5 250 500 1000
Charcoal transport (arcsine) 62.8 40.2 77.0 79.8 83.5
± ± ± ± ±
12.5 16.4# 5.75 5.30 * 7.58 **
The results are the mean ± S.E.M. ANOVA, Dunnett's Test: # p
The phytochemical analyses carried out in this work led to the isolation of xanthones and flavones in the MeOH extract of scapes from L. flavescens (Figs 1, 2). The literature reveals the gastroprotective action of various species containing xanthones and flavones (Gonzalez & Di Stasi, 2002; La Casa et al., 2000; Sartori et al., 1999; Ares et al., 1995). The flavones isolated from L. flavescens are mostly derived from luteoline which presents the catechol nucleus at ring B. Such compounds have been correlated with the antiulcerogenic activity of many plant extracts since they possess antioxidant action (La Casa et al., 2000). Moreover, the isolated xanthone 3 also displays ortho-hydroxyl groups at position 5, 6 of the A ring, thus providing another system with a catechol nucleus. Besides the presence ofxanthones and flavonoids in the MeOH extract of L. flavescens, this extract also has potential as a new alternative in the management of gastric ulcers by the presence of luteolin that has a proven effect against Helicobacter pylori (Chung et al., 2001). In light of the fact that H. pylori is considered the most important risk factor in development of duodenal and gastric ulcers, this study with MeOH extract from L. flavescens as antiulcerogenic agent may contribute an excellent pathway for treating the universal problem of this gastric pathology. In conclusion, our study evidenced the gastroprotective factor of the MeOH extract obtained from L. flavescens, with antioxidant, anti secretory and gastroprotective activities. The phytochemical investigation of the
Antioxidant Activities from Leiothrix flavescens
277
MeOH extract led to the isolation of flavones and xanthones with catechol nucleus in their structures, which can be correlated with antioxidant activity.
ACKNOWLEDGEMENTS We acknowledge the financial support ofFAPESP, CNPq and Fundunesp.
REFERENCES Agrawal, P.K. (1989). Carbon 13 NMR of flavonoids. Elsevier: Amsterdam. Ares, J.J., Outt, P.E., Randall, J.L., Murray, P.D., Weisshaar, P.S., O'Brien, L.M., Ems, B.L., Kakodkar, S.V., KeIrn, G.R and Kershaw, W.C. (1995). Synthesis and biological evaluation of substituted flavones as gastroprotective agents. Journal of Medicinal Chemistry, 25: 4937-4943. Batista, L.M., de Almeida, AB., de Pietro Magri, L., Toma, W., Calvo, T.R, Vilegas, W. and Souza Brito, AR (2004). Gastric antiulcer activity of Syngonanthus arthrotrichus Silveira. Biological & Pharmaceutical Bulletin, 27: 328-332. Bobrove, AM. (1990). Misoprostol, diarrhoea, and psyllium mucilloid. Annual International Medicine 23: 112-386. Chung, J.G., Hsia, T.C., Kuo, H.M., Li, Y.C., Lee, Y.M., Lin, S.S. and Hung, C.F. (2001). Inhibitory actions of lute olin on the growth and aryl amine Nacetyl transferase activity in strains of Helicobacter pylori from ulcer patients. Toxicol In Vitro, 15: 191-198. Coelho, RG., Batista, L.M., Dos Santos, L.C., Brito, A.RM.S. and Vilegas, W. (2006). Pythochemical study and antiulcerogenic activity of Syngonanthus b~sulcatus (Eriocaulaceae). Brazilian Journal of Pharmaceutical Science, 42: 413-417. Di Stasi, L.C., Camuesco, D., Nieto, A., Vilegas, W., Zarzuelo, A and Galez, J. (2004). Intestinal anti-inflammatory activity of paepalantine, an isocoumarin isolated from the capitula of Paepalanthus bromelioides, in the trinitrobenzenesulphonic acid model of rat colitis. Planta Medica, 70: 315-320. Dokkedal, A.L. and Salatino, A (1992). Flavonoids of Brazilian species of Leiothrix (Eriocaulaceae). Biochemistry Systematic and Ecology, 20: 31-32. Farinati, F., Cardin, R, Cassaro, M., Bortolami, M., Nitti, D., Tieppo, C., Zaninotto, G. and Rugge, M. (2008). Helicobacter pylori, inflammation, oxidative damage and gastric cancer: a morphological, biological and molecular pathway. European Journal Cancer Prevention, 3: 195-200. Fee, J.A and Teitelbaum, H.D. (1972). Evidence that superoxide dismutase plays a role in protecting red blood cells against peroxidative hemolysis. Bwchemistry Biophysic Research Communication, 49: 150-158. Fintelmann, V. (1991). Modern phytotherapy and its uses in gastrointestinal conditions. Planta Medica, 57: 48-52. Gadhi, C.A, Benharref, A, Jana, M. and Lozniewski, A (2001). Anti- Hehcobacter pylori activity of Aristolochia paucinervis pomel extracts. Journal of Ethnopharmacology, 75: 203-205. Garner, A. (1992). Adaptation in the pharmaceutical industry with particular reference to gastrointestinal drugs and diseases. Scandinavwn Journal of Gastroenterology, 27: 83-89. Giulietti, A.M., Amaral, E.C. and Biittrich, V. (1994). Phylogenetic analysis of inter and infrageneric relationships of Leiothrix Ruhl. (Eriocaulaceae). Kew Bulletin, 50: 5571. Giulietti, A.M., Pirani, J.R. and Menezes, N.L. (1998). Estudos em sempre-vivas: importancia economica do extrativismo em Minas Gerais, Brasil. Acta Botanica Brasil, 1: 179-183. Gonzalez, F.G. and Di Stasi, LC. (2002). Anti-ulcerogenic and analgesic activities ofthe leaves of W~lbrandw ebracteata in mice. Phytomedicme, 9: 125-134.
278
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Guth, P.H., Paulsen, G. and Nagata, H. (1984). Histologic and microcirculatory changes in alcohol-induced gastric lesions in the rat: effect of prostaglandin cytoprotection. Gastroenterology, 87: 1083-1090. Hachem, C.Y., Clarridge, J.E., Reddy, R., Flamm, R., Evans, D.G., Tanaka, S.K. and Graham, D.Y. (1996). Antimicrobial susceptibility testing of Helicobacter pylori. Diagnostic Microbiology Infection Disease, 41: 24-37. Harborne, J.B. (1996). The flavonoids: advances in research since 1986. Chapman & Hall: New York. Kinoshita, M., Tsunehisa, N. and Tamaki, H. (1995). Effect of a combination of ecabet sodium and cimetidine on experimentally-induced gastric-lesions and gastric-mucosal resistance to ulcerogenic agents in rats. Biological Pharmaceutical Bulletin, 18: 223-226. La Casa, C., Villegas, 1., Alarcon de la Lastra, C., Motilva, V. and Martin Calero, M.J. (2000). Evidence for protective and antioxidant properties of rutin, a natural flavone, against ethanol induced gastric lesions. Journal of Ethnopharmacology, 71: 45-53. Levine, R.J. (1971). Peptic Ulcer. Munksgard, Copenhagen, pp. 92-97. Loomis, T.A. and Hayes, A.W. (1996), Essentials of Toxicology (4th edn). Academic Press Limited: London. pp. 33-46. Mizui, T. and Doteuchi, M. (1983). Effect ofpolyamines on acidified ethanol- induced gastric lesions in rats. Japanese Journal of Pharmacology, 33: 939-945. Morimoto, Y., Shimohara, K., Oshima, S. and Sukamoto, T. (1991). Effects of the new antiulcer agent KB-5492 on experimental gastric mucosal lesions and gastric mucosal defensive factors, as compared to those of terprenone and cimetidine. Japanese Journal Pharmacology, 57: 595-605. Motilva, V., Lopez, A., Martin, M.J., La Casa, C. and Alarcon de la Lastra, C. (1996). Cytoprotective activity of cisapride on experimental gastric mucosal lesions induced by ethanol. Role of endogenous prostaglandins. Prostaglandins, 52: 63-74. Olfert, E.D., Cross, B.M. and McWilliam, A.A. (1993). Guide to the Care and Use of Experimental Animals. Canadian Council on Animal Care Co: Ontario pp. 1-213. Puscas, 1., Puscas, C., Coltau, M., Pas~a, R., Torres, J., Marquez, M., Herrero, E., Fillat, O. and Ortiz, J.A. (1997). Comparative study of the safety and efficacy of ebrotidine versus ranitidine and placebo in the prevention of piroxicam-induced gastroduodenal lesions. Arzneimittelforschung, 47: 568-572. Repetto, M.G. and Llesuy, S.F. (2002). Antioxidant properties of natural compounds used in popular medicine for gastric ulcers. Brazilwn Journal of Medicinal Biological Research, 35: 523-534. Salatino, A., Salatino, M.L.F. and Giulietti, A.M. (1990). Contents of soluble phenolic compounds of capitula of Eriocaulaceae. Quimica Nova, 13: 289-292. Santos, L.C., Piacente, S., De Ricardis, F., Eletto, A.M., Pizza, C. and Vilegas, W. (2001). Xanthones and flavonoids from Leiothrix curvifolia and Leiothrix {lavescens. Phytochemistry, 56: 853-856. Sartori, N.T., Canepelle, D., de Sousa, P.T. Jr. and Martins, D.T. (1999). Gastroprotective effect from Calophyllum brasiliense Camb. bark on experimental gastric lesions in rats and mice. Journal of Ethnopharmacology, 67: 149-156. Shay, H., Komarov, S.A., Fels, S.S., Meranze, D., Gruestein, M. and Siplet, H. (1945). A simple method for the uniform production of gastric ulceration in the rat. Gastroenterology, 5: 43-6l. Silva, M.A., Oliveira, A.P., Sannomiya, M., Sano, P.T., Varanda, E.A., Vilegas, W. and Santos, L.C. (2007). Flavonoids and a naphthopyranone from Eriocaulon ligulatum and their mutagenic activity. Chemical and Pharmaceutical Bulletin, 55: 1635-1639. Singh, S. and Majumdar, D.K. (1999). Evaluation of the gastric antiulcer activity of fixed oil of Ocimum sanctum (Holy Basil). Journal of Ethnopharmacology, 65: 13-19. Souza Brito, A.R.M. (1995). Manual de Ensaios Toxicologicos in vivo. Editora da UNICAMP, Campinas, Sao Paulo. Stickney, J.C. and Northup, D.W. (1959) Effect of gastric emptying upon propulsive motility of small intestine in rat. Proceed Society Experimental Biological Medicine, 10: 101-582.
Antioxidant Activities from Leiothrix flavescens
279
Stocks, J., Gutteridge, J.M., Sharp, R.J. and Dormandy, T.L. (1974). Assay using brain homogenate for measuring the antioxidant activity of biological fluids. Clinical Science Molecular Medicine, 47: 215-222. Szelenyi, I. and Thiemer, K. (1978). Distention ulcer as a model for testing of drugs for ulcerogenic side effects. Archieve Toxicolology, 41: 99-105. Wallace, J.L. (2001). Mechanisms of protection and healing: Current knowledge and future research. Amencan Journal Medicine, 110: 19-22.
"This page is Intentionally Left Blank"
14 Evaluation of the Immunity Potency and Security of Inactivated Oil-Adjuvant Vaccine against Chlamydiosis in Dairy Cattle Qru, C.l,*, ZHOU, J.l, CAO, X.l
AND
LIN, G.l
ABSTRACT The inactivated oil-adjuvant vaccine against chlamydiosis in dairy cattle was successfully developed using Chlamydia psittaci SX5 field strain with strong and stable virulence in China. The minimal effective dosages for adult dairy cow and calf are 3 mL and 1.5 mL, respectively, and the clinically used dosages are 4-5 mL (adult cattle) and 2 mL (calves), respectively. The average protection rate reaches 94.4% (ranging from 88.9% to 100%) in the vaccines potency tests. The immunity persistent periods of the vaccine is ten months (adult cattle) and six months (calf). The vaccines conserved for 12 months at 4°C has effects same as the new one. The safe tests for cattle of various ages and small areas of field trials (1216 cattle were vaccinated) confirmed safety of the vaccine is reliable and there were not obviously abnormal changes in appetite, milk production and the site of vaccination in the vaccinated cattle herds. Key words : Chlamydiosis in dairy cattle, inactivated vaccine, protection rate, safety
INTRODUCTION Chlamydiae are widely distributed throughout the world, causing various forms of disease in animals and humans. Several species, particularly Chlamydophila (Cp.) psittaci and Cpo abortus, are known to be transmissible 1. State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research
*
Institute, CAAS, Lanzhou Gansu 730 046, China. Corresponding author: E-mail: [email protected]
282
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
from animals to humans, causing significant zoonotic infections (Gnutov et al., 1980; Stepanek et al., 1983; Sachse et al., 2002). Chlamydiae is strictly intracellular microorganism, which have unique biphasic lifestyle, namely infective extracellular phage and parasitic intracellular phage, which renders the respective diseases difficult to control. This is compounded by the specialist growth conditions for the organisms and the lack of a genetic based system for the transformation of chlamydiae, both of which have hampered research on these pathogens. To escape the host immune response these bacteria are capable of transforming into persistent stages of development characterised by a distinct antigenic profile. Bovine chlamydial abortion is one of all serious diseases induced by Chlamydia, which may result in large loss in dairy industry (Daniel et al., 1993; Kaltenboeck et al., 2005). The disease is named as epizootic bovine abortion caused by Cp abortus, and characterized by abortion, premature birth, still birth or weak birth with wide prevalence (Martel et al., 1983; Martinov, 1984). The disease often aggrieves virgin cows, heifers or cows from nonepidemic areas and results in 25% to 75% of affected pregnant cows to abort and infertility (Bowen et al., 1978). The causative agents may induce uteritis and infertility in cows with bad curative effect despite drugs (Veznik et al., 1996), conjunctivitis (Otter et al., 2003), mastitis and polyarthritis (Martinov et al., 1981), etc. In a regional survey in Germany, Cpo psittacilCp. abortus were detected at a rate of up to 100% in affected herds. Economic losses as a consequence of a drop in milk production and milk quality, as well as abortions and reduced fertility rates were estimated to be Euro 40,000 per year at an average farm of 60 dairy cows and 20 heifers. Investigations carried out in Italy have shown that the chlamydial organisms involved in these outbreaks belong to the so-called "noninvasive" strains classified as Cpo pecorum. The same agent was also detected in cases of encephalomyelitis in water buffaloes used for milk production (Qiu, 2003). In 1980's, investigations of bovine chlamydiosis carried out in some provinces in China showed infection rate of chlamydiosis in dairy cows was higher than in yellow cattle or buffalo (Qiu, 2005). In current years, the dairy farming has been developing rapidly and at present has reached above ten million cattle in China. However, the production level of dairy cattle was very low owing to prevalence of infectious diseases (including chlamydiosis) (Qiu et al., 2006) and absence of specific bio-preparations for monitor and control of dairy cattle diseases. So we carried out studies on inactivated vaccine against chlamydiosis in dairy cattle.
Immunity of Inactivated Oil-Adjuvant Vaccine against Chlamydiosis
283
MATERIALS AND METHODS The Vaccine Strain The vaccine strain is Chlamydia psittaci SX5 which was isolated from a dairy cow affected with chlamydial abortion on a dairy farm standing at the north of Xi-an city of Shaanxi province locating in the North-western of China, and identified and preserved in our laboratory and its virulence value reached 1O-12ELD5JOA mL (Qiu et al., 2006).
The Virulent Strain Used in the Vaccine Potency Test The virulent strain used in the vaccine potency test is Chlamydia psittaci SX5 with virulence value of 1O-12ELD 5JOA mL. In the potency tests, the calves were challenged at a dosage of 1 mL and adult dairy cattle at dosage of 2 mL by neck vein route (The concentration of the virulent strain SX5 used in the vaccine potency test was 10% of suspension of the strain SX5 chicken embryo yolk sac cultures). And the challenges were carried out at the third week post vaccinations in the potency tests (Li et al., 1995).
The Tested Vaccine The ten batches of inactivated oil-adjuvanted vaccines against chlamydiosis in dairy cattle were prepared for potency test and security test of target animals and the vaccines were composed of inactivated SX5 antigens and Montanide ISA 206 oil adjuvant made in France with 0.2% formaldehyde as inactivator. In these trials, calvies and adult cattle were intramuscularly vaccinated at a doses of 2 mL and 4-5 mL, respectively.
The Vaccine Tests The contents of vaccine evaluation examination included potency test, safety test, immune persistence period test for dairy cattle and conservation time of the vaccine in the laboratory as well as field tests of vaccinations of target animals.
Animals used in the tests: All cattle were purchased from several cattle herds which had been confirmed to be negative for Chlamydia infection by serologic method.
RESULTS The Minimal Effective Dosages for Target Animal The minimal effective dosages of the vaccine for adult cattle and calf were seen in the Tables 1 and 2, respectively.
284
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Table 1. The minimal effective dosages for adult dairy cow Groups 1 2 3 4 5 6 Control
Vaccine batch no.
Cattle
030312 030317 030327 030312 030317 030327
5 5 5 5 5 5 5
Date 03-03-30 03-03-30 03-03-30 03-03-30 03-03-30 03-03-30 03-03-30
Dose
Challenge
(mL)
(mL)
Protection rate
2.0 2.0 2.0 3.0 3.0 3.0 0
2.0 2.0 2.0 2.0 2.0 2.0 2.0
2/5 2/5 2/5 4/5 5/5 5/5 0/5
Dose
Challenge
(mL)
(mL)
Protection rate
1.0 1.0 1.0 1.5 1.5 1.5 0
1.0 1.0 1.0 1.0 1.0 1.0 1.0
1/5 215 215 5/5 4/5 5/5 0/5
Table 2. The minimal effective dosages for dairy calvies Groups 1 2 3 4 5 6 Control
Vaccine batch no.
Cattle
030312 030317 030327 030312 030317 030327
5 5 5 5 5 5 5
Date 03-03-30 03-03-30 03-03-30 03-03-30 03-03-30 03-03-30 03-03-30
According to the results of minimal effective dosages tests, the minimal effective dosages of the vaccine for adult cattle and calf were 3.0 mL and 1.5 mL, respectively. Clinically, the dosages used for calves and adult cattle were affirmed as 2 mL and 4-5 mL, respectively.
The Vaccine Potency Tests Six batches of the vaccines were used for two potency tests, and in the first potency test, the vaccine protection rate was 8/9, and in the second potency test, the vaccine protection rate was 9/9, with an average protection rate of 94.4%. The detail was seen in the Table 3. Table 3. Potency tests of the vaccine for dairy cattle Groups
Vaccine Cattle batch no.
Dose (mL)
Challenge Smear Isolation Protection (mL) rate(%)
1 2 3 Control 1
030312 030327 030403
3 3 3 3
5.0 5.0 5.0 0
2.0 2.0 2.0 2.0
3/3 3/3 3/3 3/3+
3/3 3/3 2/3 3/3+
3/3 (100.0) 3/3 (100.0) 2/3 (66.7) 0/3 (0)
4 5 6 Control 2
040113 040210 040317
3 3 3 3
5.0 5.0 5.0 0
2.0 2.0 2.0 2.0
3/3 3/3 3/3 3/3+
3/3 3/3 3/3 3/3+
3/3 (100.0) 3/3 (100.0) 3/3 (100.0) 0/3 (0)
Immunity of Inactivated Oil-Adjuvant Vaccine against Chlamydiosis
285
The Safety Tests The security tests of the vaccine would deal with weaned calves (30 to 40 days old), pregnant cows and adult cattle. The details were seen in the Tables 4, 5, 6, respectively. Table 4. Safety tests of the vaccine for dairy calvies Groups 1 2 3 4 5
Vaccine batch no.
Calves
030312 030327 030403 040113 040210
5 5 5 5 5
Dose (mL)
5.0 5.0 5.0 5.0 5.0
Site of vaccination No No No No No
swelling swelling swelling swelling swelling
Appetite
Evaluation
Normal Normal Normal Normal Normal
Safe Safe Safe Safe Safe
Table 5. Safety tests of the vaccine for pregnant dairy cows Groups Vaccine Pregnant Dose (mL) batch no. cows 1 2 3 4 5
030312 030327 030403 040113 040210
5 5 5 5 5
10.0 10.0 10.0 10.0 10.0
Site of vaccination absorded in absorded in absorded in absorded in absorded in
Parturition Evaluation
3 days 2-3 days 2-3 days 2-3 days 2-3 days
Normal Normal Normal Normal Normal
Safe Safe Safe Safe Safe
Table 6. Safety tests of the vaccine for adult dairy cattle Groups 1 2 3 4 5
Vaccine Adult batch no. cows 041210 050103 050114 050120 050220
5 5 5 5 5
Dose (mL)
10.0 10.0 10.0 10.0 10.0
Site of vaccination absorded in 2-3 days absorded in 3 days absorded in 2-3 days absorded in 2-3 days absorded in 3 days
Clinically Evaluation Normal Normal Normal Normal Normal
Safe Safe Safe Safe Safe
The test results of safety of the vaccine for target animals well indicated that after the vaccinations, no tested cattle showed serious manifestations with basically normal appetite and milk crops clinically. And all of the vaccinated pregnant cattle did not aborted within the whole test duration. A few of the tested animals had a high temperature (to 39°e or so) but returned normal in the second day post the vaccinations.
The Immune Persistence Period Test of the Vaccine Nine adult cattle and nine calvies were inoculated with the vaccines and then the sera were continuously sampled from the tested animals for ten months (adult cattle) or six months (calves) for measuring the antibody levels to chlamydiosis. The antibody titer changes were seen in the Fig 1 and 2 in detail.
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
286
N
Cl
Qj
;>,
"0
10 8
6
-+- 3mL-group
4
- . - 5mL-group
0
.0
~
2 0 0
..... N
0
CD
0
(j)
0 N
0
L()
0 0 ex:> ..... ..... N
0
<;f"
N
0 ..... N
0 0
(V)
If)
;>,
co
"0
Fig 1. The antibody curves in adult dairy cattle inoculated with various doses of the vaccine within 10 months duration
As showing in the Fig 1, the antibody titers of the both group animals reached 1:16-1:32 on the day 21 post vaccination; the antibody titers of the both group animals were kept in an increasing status and reached 1: 512 on the day-90 post vaccination; the antibody titers of the both group animals were steady in 1: 256 or so within 4 to 8 months after vaccinations; in 9 to 10 months post vaccinations, the antibody level of the 3 mL-group animals began to fall but the antibody level of the 5 mL-group animals was kept in 1: 128-1: 256.
The adult cattle of the 5 mL-group were challenged with virulent strain SX5 10 months after the vaccinations and all inoculated animals were protected and the test results were seen in the Table 7 in detail.
12 10
'ON ... en
~~
~.~
'0'0 0>-
.oE ._
c:C
-+-Group I ---Group II ---.....- Group III
8 6
4
~ Control
«u 0
o
15
30
60
90
120 150 180
Days
Fig 2. The antibody curves in calvies inoculated with the vaccine within 6 months duration
Immunity of Inactivated Oil-Adjuvant Vaccine against Chlamydiosis
287
Table 7. The challenge test in the immunity persistent period of the vaccine for adult cattle Groups 1 2 3 Control
Vaccine Cattle batch no. 030312 030327 030403
3 3 3 3
Dose (mL)
Challenge Smear Isolation Protection (mL) rate(%) 3/3 3/3 3/3 3/3+
2.0 2.0 2.0 2.0
5.0 5.0 5.0 0
3/33/3 3/3 3/3+
3/3 (100.0) 3/3 (100.0) 3/3 (100.0) 0/3 (0)
15 days after the vaccinations, the antibodies (1:64) could be detected from most of the inoculated calves. And 6 months after the vaccination, the antibody titers were kept at higher levels in inoculated calves of the 3 groups and the mean antibody titers were 10 Lg2, 9.3 Lg2 and 10 Lg2, respectively. While all of the calves were challenged, the animals of the vaccinated groups all were protected (3/3, 3/3, 3/3), but no animals of the control group were protected (0/3). The test results were seen in the Table 8 in detaiL The 3/3 calves of the control group showed high body temperature (39.8°C to 40.1°C and the 1/3 calves diarrhoea occurred after challenging. Table 8. The challenge test in the immunity persistent period of the vaccine for calvies Groups
Calvies
Inoculated dose(mL)
Antibody titer 180 days post vaccination
Challenge dose(mL)
Protection rate%
I
3 3 3
2.0 2.0 2.0 3
10Lg2 9.3Lg2 lOLg2 0
1.0 1.0 1.0 1.0
3/3 (100.0) 3/3 (100.0) 3/3 (100.0) 0/3(0)
IT III Control
The conservation time of vaccine The three batches of the vaccine which had been hold for 12 months at 4°C were used to inoculate 9 dairy cattle, each team 3 animals. Challenging on the day 21 post the vaccinations, all of the vaccinated animals were protected but the 3 control animals fell ill showing high body temperature, bad appetite or abortion. The test results were seen in the Table 9. Table 9. Storage period tests of the vaccine Groups 1 2 3 Control
Vaccine Cattle Vaccination batch no. date 030312 030327 030403
3 3 3 3
04-04-05 04-04-05 04-04-05 04-04-05
Dose (mL)
Challenge date
5.0 5.0 5.0 0
04-04-26 04-04-26 04-04-26 04-04-26
Dose Protection (mL) rate% 2.0 2.0 2.0 2.0
3/3 (100.0) 3/3 (100.0) 3/3 (100.0) 0/3(0)
288
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
According to the test results, the quality of the vaccines kept for 12 months at 4°C did not go down. So the conservation time of the vaccine was a year.
The Vaccine Field Test In order to further know effect and safety of the vaccine for dairy cattle in field, the three batches of the vaccine were used in field tests and 1216 dairy cattle were vaccinated at some of the dairy farms in Gansu, Ningxia, Inner Mongolia, Shaanxi, Henan, Guangdong, Zhejiang and Sichuan provinces, China, which proved that the vaccines were effective for preventing and controlling bovine chlamydiosis and very safe. For example, in 2003 year, on a dairy farm with above one thousand cattle, six hundred of which became pregnant in Ningxia province, chlamydiosis took place,which induced above 100 pregnant cows to abort within a month with suffering heavy economic loss. Mterwards, when all animals of the herd were vaccinated with the vaccines, abortions of dairy cows were stopped rapidly and the production of the farm recovered normality also. DISSCUSSION Bovine chlamydial abortion is one of chlamydiosises leading to very severe economic loss in dairy farming in China. Now the natural transmission manner of the disease has been unclear. The affected animals and latently infected hosts carrying the pathogen and showing no clinical manifestations were very important infection origins (Reinhold et al., 2008). It might be possible that the disease was transmitted between cattle and sheep each other (Griffiths et al., 1995). While pregnant cows with chlamydiosis aborted or laboured, a lot of Chlamydia would be discharged out with the amniotic fluids (Wittenbrink et al., 1994) and polluted environment around and infected other susceptible animals via ingestive tracts (Wittenbrink et al., 1993) and respiratory tracts (Ehret et al., 1975). If a breeding bull was infected by Chlamydia psittaci, cow herds copulated with the bull might be infected by Chlamydia- contaminated sperm (Storz et al., 1968; Amin 2003; Kauffold et al., 2007). Investigations revealed that if not controlling, bovine chlamydiosis would be rapidly spread and induce very severe economic loss in cattle farming (Stepanek et al., 1983). Because of extensive occurrences and many infection routes of Chlamydia spp, the complex strategy characterized by vaccination and strong management would be needed for prevention and control of chlamydiosis in cattle (Rodolakis et al., 1987; Rodolakis et al., 1998; DeGraves et al., 2004; Biesenkamp-Uhe et al., 2007). The very pivotal aspect for developing a vaccine is selection of immunogenecity-broad, virulence-steady vaccine strain. In the study, C. psittaci SX5 was confirmed to be virulence-strong and stable by continuous passages in chick embryos, clone and sequence of the MOMP gene and
Immunity of Inactivated Oil-Adjuvant Vaccine against Chlamydiosis
289
potency test of target animal and highly identical in homology of the MOMP genes of prevalence strains from other regions (Qiu et al., 2006). So the SX5 is a more ideal strain for vaccine. According to minimal vaccination dosage test, the dosages used clinically for calvies and adult cattle were 2 mL and 4-5 mL, respectively. The dosages for adult dairy cows would be chosen on the basis of the milk production and individual bodily form size, if the milk production was higher and the individual bodily form size was bigger, the vaccination dose would be 5 mL, otherwise, 4 mL. In the security tests, because two-months-pregnant cows were easily infected with Chlamydia sp the safety of the vaccine to two-monthspregnant cows was examined which showed that the vaccine was much safe for both mother and fetus. So the efficacy of vaccinations for preventing chlamydial infection in dairy cows would be reasonable in a month before or after artificial inseminations. On the basis of prevalence characteristics of bovine chlamydiosis and duration of the vaccine protection, the use procedure of the vaccine: cows will be vaccinated in a month before or after artificial inseminations; calves will be vaccinated in a month after weaning.
ACKNOWLEDGEMENTS This research was supported by The director funds of Lanzhou Veterinary Research Institute (No. 2003-6; No. 2005-5); The national specific fund of dairy industry (No. 2002BA518A04); The national science-technology support plant fund (No. 2006BAD 04A05) and the Gansu province grand specific project fund of biologic technique (No. 0702NKDA037).
REFERENCES Amin, A.S. (2003). Comparison of polymerase chain reaction and cell culture for the detection of Chlamydophila species in the semen of bulls, buffalo-bulls, and rams. Vetennary Journal, 166(1): 86-92. Biesenkamp-Uhe, C., Li, Y., Hehnen, H.R., Sachse, K. and Kaltenboeck, B. (2007). Therapeutic Chlamydophila abortus and C. pecorum vaccination transiently reduces bovine mastitis associated with Chlamydophila infection. Infection and Immunity, 75(2): 870-7. Bowen, RA., Spears, P., Stotz, J. and Deidel, G.E. Jr. (1978). Mechanisms of infertility in genital tract infections due to Chlamydia psittaci transmitted through contaminated semen. The Journal of Infective Diseases, 138(1): 95-8. Daniel, RG., Holliman, A., David, G.P., Kirby, F.D., Simpson, V.R, Cranwell, M.P., Dawson, M. and Griffiths, P.C. (1993). Bovine chlamydiosis in the United Kingdom. The Veterinary Record, 133(14): 351-35. DeGraves, F.J., Kim, T., Jee, J., Schlapp, T., Hehnen, H.R and Kaltenboeck, B. (2004). Reinfection with Chlamydophila abortus by uterine and indirect cohort routes reduces fertility in cattle preexposed to Chlamydoph~la. Infectwn and Immunity, 72(5): 2538-45. Ehret, W.J., Schutte, A.P., Pienaar, J.G. and Henton, M.M. (1975). Chlamydiosis in a beef herd. Journal of the South Afncan Veterinary Association, 46(2): 171-179.
290
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Gnutov, LN. and Erokhina, S.G. (1980). Clinico-serologic study of humans with diseases epidemiologically related to halproiosis(chlamydiosis) of farm animals. Zhurnal Mikrobiologii, Epidemiologii, IImmunobiologii, (10): 26-31 (in Russian). Griffiths, P.C., Plater, J.M., Martin, T.C., Hughes, S.L., Hughes, KJ., Heminson, R.G. and Dawson, M. (1995). Epizootic bovine abortion in a dairy herd: characterization of a Chlamydia ps!ttac! isolate and antibody response. The British Veterinary Journal, 151(6): 683-93. Kaltenboeck, B., Hehnen, H.R. and Vaglenov, A. (2005). Bovine Chlamydophila spp. infection: do we underestimate the impact on fertility? Veterinary Research Communications, 29(Suppl 1): 1-15. Kauffold, J., Henning, K, Bachmann, R., Hotzel, H. and Melzer, F. (2007). The prevalence of chlamydiae of bulls from six bull studs in Germany. Animal Reproduction Science, 102(12): 111-21. Li, Y., Shuai, Y., Duan, Y., Gao, S., Yang, X.J., Qiu, C., Yang, X.L., Yi, Q., Zheng, L., Chen, Z., Zhou, G., Pang, B., Liu, Q., Wei, Q. and Li, Z. (1995). Development of inactivated vaccine against chlamydial abortion in pigs. Chinese Journal of Veterinary Science and Technology, 25(11): 3-7 (in Chinese). Martinov, S., Popov, G. and Panova, M. (1981). Chlamydial polyarthritis in calves. Veterinarno- Meditsinski Nauki, 18(3): 83-91 (in Bulgarian). Martel, J.L., Perrin, M., Rodolakis, A., Russo, P., Deschanel, J.P. and Garnier, F. (1983). Experimental infection of the pregnant cow with Chlamydia psittaci. Annals of Veterinary Research, 14(2): 117-20 (in French). Martinov, S. (984). Chlamydial infection in herds of cattle with abortions. VeterinarnoMeditsinski Nauki, 21(10): 81-8 (in Bulgarian). Otter, A., Twomey, D.F., Rowe, N.S., Tipp, J.W., McElligott, W.S., Griffiths, P.C. and O'Neill, P. (2003). Suspected chlamydial keratoconjunctivitis in British cattle. The Veterinary Record, 152 (25): 787-8. Qiu, C. (2003). Chlamydiosis in animals and COST study. Infornations on Animal-Veterinary Science and Technology, 19(8): 21-24 (in Chinese). Qiu, C. (2005). Bovine chlamydiosis and its control. China Animal Health, 3: 18-20 (in Chinese). Qiu, C., Zhou, J., Cheng, S. and Cao, X. (2006). Identification and immunogenicity tests of pathogen causing chlamydial abortion in dairy cows. Vetermary Science in China, 36(4): 270-273 (in Chinese). Qiu, C., Hou, Y., Zhou, J., Cao, X. and Zhang, F. (2006). Epidemiological investigation of chlamydiosis in dairy cattle in China. Indwn Journal of Animal Scwnces. 76(12): 985-987. Reinhold, P., Jaeger, J., Liebler-Tenorio, E., Berndt, A., Bachmann, R., Schubert, E., Melzer, F., Elschner, M. and Sachse, K. (2008). Impact of latent infections with Chlamydophila species in young cattle. Veterinary Journal, 175(2): 202-11. Rodolakis, A., Salinas, J. and Papp, J. (1998). Recent advances on ovine chlamydial abortion. Veterinary Research, 29(3-4): 275-288. Rodolakis, A. and Souriau, A. (1987). Vaccination against bovine chlamydial abortion with a temperature- ensitive mutant of Chlamydia psittaci. Annals of Veterinary Research, 8(4): 439-41 (in French). Sachse, K and Grossmann, E. (2002). Chlamydial diseases of domestic animals-zoonotic potential of the agents and diagnostic issue. Deutsche tieriirztliche Wochenschift, 109(4): 142-8 (in German). Stepanek, 0., Jindrichova, J., Horacek, J. and Krpata, V. (1983). Chlamydiosis in cattle and in man: an epidemiologic and serologic study. Journal of Hygiene, Epidemiololy, Microbiology and Immunology, 27(4): 445-59. Storz, J., Carroll, E.J., Ball, L. and Faulkner, L.C. (1968). Isolation of a psittacosis agent (Chlamydia) from semen and epididymis of bulls with seminal vesiculitis syndrome. American Journal of Veterinary Research, 29(3): 549-55. Veznik, Z., Kummer, V., Canderle, J., Maskova, J., Svecova, D., Pospil, L., Diblikova, L and Zraly, Z. (1996). Detection of Chlamydia in inflammation of the cervix and uterus in cows. Veterinarnl Mediclna (Praha), 41(10): 297-304 (in Czech).
Immunity of Inactivated Oil-Adjuvant Vaccine against Chlamydiosis
291
Wittenbrink, M.M., Kirpal, G., Thiele, D., Fische,r D., Krauss, H. and Bisping, W. (1994). Detection of Chlamydia psittaci in vaginal discharge of cows: a necessary enlargement of bacteriologic diagnosis for the etiologic clarification of fertility disorders in the female cow. Journal of Veterinary Med~clne Series B, 41(7-8): 492-503 (in German). Wittenbrink, M.M., Bisping, W., Mrozek, M. and Horchler, H. (1993). Intestinal Chlamydia psittaci infection of cattle: frequency and technical aspects of the cultural detection of the agent. Deutsche tierarztliche Wochenschift, 100(5): 195-8 (in German).
"This page is Intentionally Left Blank"
15 Annona crassiflora Wood Constituents: Antimalarial Efficacy, Larvicidal and Antimicrobial Activity MARy Am; GoNQALVESl, VANESSA CARLA FURTADO MOSQUEIRA2 AND LUCIA PINHEIRO SANTOS PIMENTA 1,*
ABSTRACT Wood of Annona crassiflora was phytochemically evaluated and three annonaceous acetogenins, annofolin (1) ACM5 (2) and ACM6 (3), the alkaloids aterospermidine (4) and liriodenine (5), the steroids f3-sitosterol (6) and stigmasterol (7), and fatty acids were isolated. Their structures were elucidated by a combination of chemical and spectral methods including MS and NMR experiments. Antimalarial activity, antimicrobial activity in vitro against pathogenic bacteria and brine shrimp lethality of wood extracts of Annona crassiflora were evaluated. The crude extract and fractions were very toxic to brine shrimp (DL 50<30 J.1g/mL). Doses of the extract and fractions of 500 mg/kg and 250 mg/kg caused animal death upon injection in P.berghei-infected mice. Doses of 12.5 mg/kg and 1.25 mg/kg produced 67% suppression of parasitaemia. The alkaloidic fraction showed good results when compared with chloramphenicol against pathogenic bacteria. Key words: Annona crassiflora, Annonaceae, antibacterial activity, antimalarial efficacy, larvicidal activity, Plasmodium berghei
1. Departamento de Quimica - Instituto de Ciencias Exatas - Universidade Federal de
Minas Gerais, Av. Antonio Carlos 6627, BeloHorizonte, MG, Brazil; 31270-901. 2. Departamento de Farmacia- Escola de Farmacia - Universidade Federal de Duro Preto, Duro Preto, MG. * Corresponding author: E-mail: [email protected]
294
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
INTRODUCTION It is estimated that Brazil has about 60-250000 plant species that still are lesser known and under explored. Probably, 40% of them possesses interesting biological activities, constituting source of new useful molecular standards for the development of new plant-derived drugs. Our team has been investigating Annonaceae family for over ten years (Santos et al., 1995; Pimenta et al., 1996; Pimenta et al., 2001; Santos et al., 2006) to find plants with potential activity as antimicrobial, against endemic illnesses such as malaria, and chronic-degenerative diseases as the cancer. Annonaceae family has been source of several structural types of isoquinoline alkaloids and Annonaceous acetogenins. Annonaceous acetogenins are long chain fatty acid derivatives isolated exclusively from plants belonging to the Annonaceae family. These natural products exhibit a broad range of biological properties such as cytotoxic, immunosuppressive, pesticidal, antiparasitic, and antimicrobial activities. Furthermore, its potential to inhibit multiple drug resistant cells have attracted increasing interest in these compounds (Bermejo et al., 2005). Our previous phytochemical study with the seeds of Annona crassiflora has yielded three new and four known annonaceous acetogenins that showed good cytotoxic activity against some human tumoral cells lines (Santos et al., 1995; Pimenta et al., 1996; Pimenta, 1995). Insecticidal activity against Spodoptera frugiperda was also detected from leaves and seeds extracts of Annona crassiflora (Pimenta et al., 2000). The Annonaceous acetogenins had also been determined earlier to possess antiparasitic activities, such as antimalarial in vivo (Rupprecht et al., 1990; Fang et al., 1993). Isoquinoline alkaloids have shown great activity against resistant strains of P. falciparum. (Fisher et al., 2004). Xylopia aromatica bark extract (Annonaceae) was active against Plasmodium falciparum chloroquine resistant strain in vitro but inactive against P. berghei ANKA strain in vivo (Garavito et al., 2006). Considering that malaria is the most prevalent disease among the insectborne individuals and kills around two million people every year, it is very important to look for antimalarial active new drugs. The success of the antimalarial drugs, quinine and artemisinin, from plant's sources, associated to the ethnopharmacological information about the Xylopia aromatica activity in vitro against P. falciparum (Garavito et al., 2006) and also the presence of the acetogenins and isoquinoline alkaloids in this species drove our attention to the evaluation of the antimalarial activity of the wood's crude extracts of A. crassiflora and some fractions from them. The species Annona crassiflora, popularly known as "araticum, cabe~a de-negro", or "marolo", is a native tree from Brazilian "cerrado". It ocurrs in the states of Minas Gerais, Bahia, Ceara, Goias, Mato Grosso and Sao Paulo. Its fruit is edible and used as juice, ice-cream, jelly and liqueur. Its pulp is laxative and the seeds are used against diarrhoea and once pulverized they are toxic and reputed as anti-parasitic. It has been used as efficient
Annona crassiflora Wood Constituents
295
remedy for lice elimination. There are reports of leaves being used as medication for sweat, antirheumatism by oral route. It may also be used externally, as a mouth rinse, in treatment of stomach inflammations, and in neuralgias and headaches (Lorenzi et al., 2002). Prior to our investigation some alkaloids have been isolated from the leaves (Hocquemiller et al., 1982), and cytotoxic annonaceous acetogenins from the seeds (Santos et al., 1994; Santos et al., 1999; Pimenta, 1995) of this plant.
Annona crassiflora is disappearing due to the fire used by local farmers for preparing the field for crops. Neither chemical nor biological studies of this plant's wood are reported in the literature. The present paper reports the fractionation of A. crassiflora wood extract bioguided by Artemia salina lethality test which lead to the isolation of three annonaceous acetogenins, named annofolin, ACM5 and ACM6 (1, 2, 3), the alkaloids aterospermidine (4) and liriodenine (5), the steroids psitosterol (6) and stigmasterol (7), and fatty acids. The antimalarial efficacy and the general toxicity of the extracts and some fractions were evaluated in vivo in the Plasmodium berghei-infected mice following the conventional four-day-test (Peters et al., 1986). Antimicrobial activity was determined in vitro against pathogenic bacteria S. aureus, B. subtilis, E. coli, M. luteus and P. aeruginosa (Takahashi et al., 2006).
MATERIALS AND METHODS Melting points determinations were determined on a Mettler FP5 apparatus and are uncorrected. Optical rotations were taken on a PERKIN ELMER 341 polarimeter. IR spectra were measured on a Perkin Elmer spectrometer version 3.02.1 and Spectrum One-FT-IR Spectrometer Universal ATR Sampling Accessory. IH NMR (400 MHz) and 13C NMR (100 MHz) spectra (all in CDCI3) were obtained in BRUKER DPX-400 spectrometer. ESIMS was recorded in a MICRO TM Q-TOF MICROMASS spectrometer. HPLC was carried out with WATERS HPLC instrument with a UV-VIS 486 detector set at 220 nm, using a Shim-pack PREP-ODS C-18 column (4.6 mm i.d. x 250 mm). Control of the equipment, data acquisition, processing, and management of chromatography information were performed by the WATERS 746 integrator. Chloroquine diphosphate was purchased from Sigma-Aldrich, USA. N,N-dimethylacetamide and PEG 300 were provided by Vetec. The solvents were from Quimex, Vetec, Carlo Erba and J. T. Baker.
Plant Materials The pieces of wood used in the present work were collected from the protected area inside of Zoo-Botanic Foundation of Belo Horizonte, Belo Horizonte, Minas Gerais state, Brazil. Voucher specimens (No. 22988) were deposited at the Instituto de Ci€mcias Biol6gicas Herbarium (BHCB), UFMG, Belo Horizonte, MG, Brazil.
296
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Extraction and Isolation The wood ofA. crassifZora was dried at 40°C, weighted (1166.00 g), grounded and extracted at room temperature with ethanol which was removed under vacuum to give the crude extracts (ACWF01; 150.9 g). Part of this extract (130.60 g) was dissolved in ethanoVwater (7:3) and successively extracted with hexane and chloroform. Mter solvent removal, the hexane (ACWF02), chloroform (ACWF03) and hydroalcoholic (ACWF04) fractions were obtained. The fraction ACWF03 was extracted with acid aqueous solution pH 4 followed by chloroform resulting in organic layer (ACWF03 0) and an acid aqueous layer (ACWF03 A). The acid aqueous phase was alkalinized and extract with chloroform, which after solvent removal yielded an alkaloidic fraction (ACW A). The ACWF03 0 was partitioned between hexane and MeOH-H 20 (9:1), furnishing the hydroalcoholic (ACWF05) and hexane (ACWF06) fractions. The ACWF05 0 fraction was submitted to biological guided fractionation on silica gel, followed by gel filtration on Sephadex LH-20 and preparative reverse phase HPLC with acetonitrilewater gradients, furnishing the acetogenins 1 (2.0 mg), 2 (2.0 mg), and 3 (1.5 mg). The alkaloidic fraction (ACW A) was fractionated on silica gel column affording the alkaloids 4 (3.0 mg), and 5 (2.5 mg). The ACWF02 fraction after separation by silica gel column chromatography eluted with hexane, dichlorometane, acetyl acetate and methanol gradients afforded the compounds 6 and 7 (7.0 mg) as a mixture.
Biological Evaluation Brine Shrimp Test Artemia salina encysted eggs (10 mg) were incubated in 100 mL of seawater under artificial lighting at 28°C, pH 7-8. After incubation for 24 h, nauplii were collected with a Pasteur pipette and kept for an additional 24 h under the same conditions to reach the metanauplii stage. The samples (triplicate) to be assayed were disolved in dimethylsulfoxide (DMSO) (2 mg/400 ~ or 2 mg/1000 ilL) and serially diluted (10, 20, 30 and 50 !Jll5mL) in seawater. About 10-20 nauplii were added to each set oftubes containing the samples. Controls containing 50 ilL of DMSO in seawater were included in each experiment. As a positive control, lapachol dissolved in DMSO was used. Twenty four h later, the number of survivors was counted, recorded and the lethal concentration 50% (LC 50 ) and 95% confidence intervals were calculated by Probit analysis (Finney, 1976). Antibacterial Assay The following bacterial strains were used: Bacillus subtilis ATCC 6633, Escherichia coli ATCC 25922, Micrococcus luteus ATCC 9341, Pseudomonas aeruginosa ATCC 27835 and Staphylococcus aureus ATCC 25923. They were cultured on Miller-Hinton agar at 37°C for 48 h. Antibacterial activity was assayed by agar diffusion method (Bauer et al., 1966). Dimethylformamide
Annona crassiflora Wood Constituents
297
(DMF) was used for extracts solubilization. Sterile 6 mm diameter filter paper discs were impregnated with 1 mL of each extract (100 llg/mL). Inhibition zones surrounding the discs were measured after 24 and 48 h of bacterial growth. Chloramphenicol (30 J..lg/disc) was used as positive control and discs containing only DMF were used as negative control. Inhibition zone diameters around each disc (diameter ofinhibition zone minus diameter of the disc) were measured and recorded at the end of incubation time. The experiment was carried on in triplicate and an average zone was calculated from all measurements. Only inhibition zones equal or bigger than 7 mm (observed after 24 h) were considered of interest.
Antimalarial Activity in P. berghei-Infected Mice. Animal experiments were carried out according to the Principles of Laboratory Animal Care and legislation in force in Brazil and the protocol was approved by the animal ethical committee of the University. Female outbred Swiss albino mice weighing 20 to 24 g were supplied by the Animal Facility of Universidade Federal de Ouro Preto. They were kept in a normal diurnal cycle and had free access to food and water throughout the experiments. The classical 4-day suppressive test adapted from Peters et al. (1986) for determination of antimalarial activity against chloroquine sensitive Plasmodium berghei NK65 strain in Swiss albino mice was used for monitoring in vivo activity of the extracts and fractions. An infective inoculum was prepared from a previously infected donor mouse with rising parasitemia (20%). On day 0 the mice were infected intravenously with a million of infected erythrocytes of P. berghei in 0.2 mL of phosphate-buffered saline. They were randomly divided in groups of 5 mice and treated by the intraperitoneal route once daily (1.25; 12.5, 250 and 500 mg kg-I) with different extracts or fractions of A. crassiflora for four consecutive days (days 0 to 3). The extracts and fractions were dissolved in N,Ndimethylacetamide and polyethyleneglycol (PEG 300) at 1:2 proportion and further diluted 15-fold in saline to obtain the desired concentration in 0,3 mL for injection. Control groups received 0.2 mL of phosphate-buffered saline (untreated) or chloroquine diphosphate (60 mg/kg/day) as positive control. Thin blood smears were made from tail blood on days 4, 7, and 16 after infection. The parasitemia levels were assessed on Giemsa-stained thin blood smears. At least 3000 cells were checked to calculate parasitemia percent. Drug activity was determined on the basis of average parasitemia per group of mice. The percent reduction of parasitemia in treated groups as compared to untreated groups was calculated as follows: % parasitemia in control group (Pcg) - % parasitemia in test group/Pcg x 100.
Statistical Evaluation All RBC counts and parasitemia levels are expressed as mean values ± standard deviations. The parasitemia data were analysed by using the one-
298
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
way analysis of variance (ANOVA) test using Prisma® 4.0 software. Mean survival times were compared by using Student's t test, considering a probability of 5% to be significant. RESULTS AND DISCUSSION
Isolation & Characterization The molecular formula of 1 was determined as C37H6607 from the [M+H]+ peak at mlz 623.53 in the electro spray ionization mass spectrum (ESIMS). A positive Kedde test (Alali et al., 1999) and IR absorptions at 3400, 2920, 2850,1745,1650 cm-l suggested 1 to be an annonaceous acetogenin bearing an a,~-unsaturated y-Iactone moiety. The presence of a typical a, ~ unsaturated-y-Iactone, without an hydroxyl group at C-4 (Alali et al., 1999; Kim et al., 2001) was suggested by the signals at 0 (H) 6.98 (1 H, td, J H-35/ H-36 = J H-35/H-3 = 1.40 Hz, H-35); 4.99 (1 H, qdd, J H-36/H-37 = 6,80 Hz; J H-35/ H-36 = J H-36/H-3 = 1.40 Hz, H-36); 1.41 (1 H, d, J H-36/H-37 = 6.80 Hz, H-37) e 2.26 (2 H, tt, J H-3/H-4 = 7.6 Hz; J H-3/H-35 = J H-3/H-36 = 1.4 Hz, H-3) in the lH_ NMRspectrum (Table 1), which showed correlations with carbons in the HMQC and HMBC spectra, permitting the assignment of carbons signals o (C) 173.94 (C-1), 148.91 (C-35), 134.40 (C-2), 77.42 (C-36) and at 19.20 (C-37). The presence of an adjacent bis-THF rings system with two flanking OH groups was suggested by the four carbon signals between 0 83.30 and 82.14, which were correlated to the proton signals at 0 3.91 (5H) in the HMQC spectrum. The one-bond lH_13C correlations detected in the HMQC spectrum, along with those observed in COSY, permitted the assignment ofthe signals at 0 74.00 and 71.80 to oxygenated methine carbons and at 0 3.42 and 03.91 to the hydrogen adjacent to the THF-rings. COSY spectrum showed correlation between these signals at 0 3.91 (5H) and the multiplet at 0 3.42 (lH). The presence of a third hydroxyl group in the hydrocarbon chain was suggested by the signals at 0 3.60 (1H) and 0 71.40 (in lH NMR and l3C NMR spectra, respectively). The placement of THF rings and the additional hydroxyl group (0 3.60 and 071.40) was determined by a close examination of the ESIMS fragmentation of 1 after induced collision of the ion at mlz 623.53. The hydroxyl group was located at C9 on basis of fragments at mlz 439, 411 e 181 in ESIMS of 1 (Figs 1,2). Only the relative stereochemistry at C-11/C-12, C-15/C-16 and C-19/C-20 was determined by comparing the lH_ and l3C-NMR data to those of model compounds of known relative stereochemistry (Cave et al., 1997; Jossang et al., 1990; Sahai et al., 1994). This comparison showed the relative configurations between the carbons C-11 to C-20 being as threo-trans-threo-trans-erythro considering the similarity ofthe lH_ and l3C-NMR data of 1 with those of the bullatacin-type of acetogenins (Zeng et al., 1996; Sahai et al., 1994). The planar structure of this adjacent bis-THF acetogenin annofolin (1) with a hydroxyl group in C-9, to the best of our knowledge, was described by our
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
300
Fig Contd.
22
~
23
6
7
Fig 1. Structures of the compounds isolated from Annona crassiflora wood
The planar structure of the ACM5 (2) is similar to annofolin (1) differing only in the relative stereochemistry around the THF rings that was determined as threo-trans-threo-trans-threo by IH NMR data (Table 1). Table 1. 1Ha and 13C NMR spectral data of anofolin (1), and 1Ha NMR data of ACM5 (2) and ACM6 (3) Annofolin
Carbon IH(J)
1 2 3 4 5-7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24
25-33 34 35 36 37
2.26 tt (7.60, 1.40) 1.54m 1.26-1.30 m 1.40m 3.60m 1.40m 3.42m 3.91m 1.54-2.00 1.54-2.00 3.91m 3.91m 1.54-2.00 1.54-2.00 3.91m 3.91m 1.40m 1.26m 1.26-1.30 m 1.26-1.30 m 1.26-1.30m 0.89 t (6.60) 6.98 d (1.40) 4.99 qd (6.80, 1.40) 1.41 d (6.80)
a CDCl3, 400 MHz. (J in Hz)
13C 173.9 134.4 25.0 27.0 22.5-29.5 32.0 71.4 32.0 74.0 83.2 27.0 28.8 82.0 82.0 28.8 28.3 82.7 71.8 32.0 22.5-29.5 22.5-29.5 22.5-29.5 22.5-29.5 14.0 148.9 77.4 19.2
ACM5
ACM6
IH(J)
IH(J)
2.26 tt (7.66, 1.52) 1.58m 1.26m 1.40m 3.60m 1.40m 3.41m 3.86-3.91 m 1.54-2.00 1.54-2.00 3.86-3.91m 3.86-3.91m 1.54-2.00 1.54-2.00 3.86-3.91 m 3.41m 1.40 1.26m 1.26m 1.26m 1.26m 0.89 t (6.40) 6.98 d (1.52) 4.99 qd (6.80, 1.52) 1.41 d (6.80)
2.26 tt (7.66,1.00) 1.56m 1.26m 1.26m 1.26m 1.26m 1.26m 1.39m 3.61m 1.39m 3.47m 3.93m 1.56-1.97 m 1.56-1.97 m 3.93m 3.93m 1.56-1.97m 1.56-1.97 m .93m 3.93m .26-1.39 m 0.88 t (6.80) 6.99 d (1.00) 4.99 qd (6.40, 1.00) 1.41 d (6.40)
Annona crassiflora Wood Constituents
301
+
37
3
o _____ ..J
(227)
~
-H20
209
Fig 2.
(367) +-H20
349
-----, -3H 0
(441)~ 387
~ -H2
439
Diagnostic ESIMS fragment ions (m/z) for annofolin (1). Peaks in parentheses were not observed
The planar structure of ACM6 (3) was suggested based on the IH_ NMR (Table 1), COSY and ESI mass spectra. The signals at 86.99 (lH, d, H35); 4.99 (lH, qd, H-36); 1.41 (3H, d, H-37) and 2.26 (2H, tt, H-3), indicated the presence of a a, ~-unsaturated-y-Iactone , without an hydroxyl group at C-4 (Alali et al., 1999). The signals at 8 3.47 (lH, m, H-15); 3.61 (lH, m, H-13) e 3.93 (5H, m, H-16, 19,20,23 e 24) are characteristics of de hydrogens of oxygenated carbons, with the signal at 8 3.61 assigned to a hydroxyl group in the hydrocarbon chain (Hisham et al., 1991) and the others assigned to a bis-THF system with two flanking hydroxyl groups. The relative stereochemistry from C-15 to C-24 was suggested as threo-trans-threo-transerythro considering the similarity of the 1H - and 13C-NMR data of 3 with 1 and those of the bullatacin-type of acetogenins (Zeng et al., 1996). The ESIMS data revealed the presence of one acetogenin with molecular formulae of C37H6607. Based on analysis of fragment ions obtained after induced collision ofthe ion mlz 623.40 [M + Hj+ the the system bis-THF a, a'-dihydroxylated was located between C-15 and C24, and the third hydroxyl group at C-13 (Fig 3). The structures of the alkaloids aterospermidine (4) and liriodenine (5) and the steroids ~-sitosterol (6) and stigmasterol (7), were confirmed by one and bidimensional IH_ and 13C-NMR data and MS measurements, compared to those of the literature (Guinadeau et al., 1975; Hossain et al., 1995; Castro et al., 1996; Goulart et al., 1993). This is the first report of the alkaloid presence in the wood of Annona crassiflora, and the first report of aterospermidine in this species.
302
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I 363
293
t-
t-
H2O
(381)
265
t
H2O
(311)
-H2
(267)
237
,r----,
23
OH
'D y5 '
"""'20! 19
0"
37
: ,, ,
0\\\
\".,
yl3 !.(CH 2 :
16
:
, ,,
I
OH
I
OH:
o
(311)
+-H,O 293
Fig 3. Diagnostic ESIMS fragment ions (m/z) for annofolin (3). Peaks in parentheses were not observed
Biological assays The crude extract (ACWF01) and the fractions ACWF02, ACWF03, ACWF03 0, ACWA, ACWF05, ACWF06 showed good bioactivity in brine shrimp lethality test with the DL50 range from 1.29 to 30 J..I.g/mL (Table 3), This is in accordance with the presence of annonaceous acetogenis as well as aporfine alkaloids.
Plasmodium berghei NK-65 strain, sensitive to chloroquine (Peters et al., 1986), was used for antimalarial evaluation in vivo. This strain is known to induce high mortality in mice, providing a good model to estimate survival and antimalarial efficacy in reducing parasitemia, and is sensitive to all currently used antimalarial drugs. It was kindly supplied frozen in nitrogen by Prof. E.M. Braga, Malaria Laboratory, Parasitology Department, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil. The antimalarial activity was determined using the four-day suppressive test in P. berghei infected mice. Doses of the extract and fractions of 500 mg/kg, 250 mg/kg, 12.5 mg/kg and 1.25 mg/kg were tested. The crude extract caused 100% of death in a dose of 500 mg/kg, and 60 % of death with 250 mg/kg in mice, by intraperitoneal route in the first 24 h after administration. Table 2 summarizes the effects of extracts and fractions of A. crassiflora at concentrations of 1.25 and 12.5 mg/kg/day in vivo. The results showed that alkaloidic fraction (ACWA) has a significant antimalarial activity compared with untreated control (p<0.05) and a mean survival time
Annona crassiflora Wood Constituents
303
greater than the control indicating the smaller toxicity of this fraction. However, the other fractions especially hexane fraction (ACWF02) exhibits higher toxicity even at the lower doses used in this protocol. In general, the percentage of parasitemia reduction increases with the dose and particularly with the time, suggesting that the pharmacokinetic profile ofthe substances responsible for the activity possesses long half-lives in mice. No fraction or extract was able to suppress 100% parasitemia (Table 2). However all the extracts and fractions produced some activity reducing the parasitaemia in 13 to 79% range. As expected the P. berghei NK65 strain was chloroquinesensitive parasite and 60 mg/kg/day had a suppressive activity of 100%. The solvents DMAIPEG/saline had no significant effect (p>0.05) on parasitemia and on the mean survival time (data not shown) compared to controls. In the antibacterial activity test by disc diffusion, the alkaloidic fraction, ACW A, showed good results against pathogenic bacteria Staphylococcus aureus, Streptococcus pyogenes, Salmonella typhimurium and Escherichia coli (Table 3). This is the first report of phytochemical contituents of Annona crassiflora wood with evaluation in different biological activity tests. The results indicated this plant as a potential source of lead compounds for the development of antitumoral, pesticide, antimicrobial and antimalarial drugs. Table 2. Mean Parasitemia (%), mean survival time and parasitemia reduction (%) in P. berghei NK65 infected mice treated with extracts and fractions of Annona
crassiflora Codes
Fractions
Dose mgIKgI day
MST# Days ±SD
Mean Parasitemia(%)# D4
Parasitemia Reduction
D7
(%)
(days 417) Control Not treated Chloroquine
16.2 ± 3.3 >30 14.3 ± 4.0§
ACWF01
60.0 Crude extract 1.25
ACWF01 ACWF02
Crude extract 12.5 Hexanic 1.25
ACWF02
Hexanic
ACWF05
Hydroalcoholic 1.25 Hydroalcoholic 12.5
13.3 ± 4.8§
Alkaloidic
1.25
20.8 ± 4.8§
Fraction from ACWF030
1.25
14.3 ±
ACWF05 ACWA F1-G24 F1-G24
12.5
Fraction from 12.5 ACWF030
14.4 ± 3.5 0 12.6 ± 1.7§
24.6 ± 3.3 0 15.9 ± 0.7*§ 8.9 ± 1.7*§ 11.4 ± 0.7*§
14.8 ± 4.4 9 13.3 ± 2.1§ 11.6 ± 2.0§ 10.2 ± 7.8§ 11.1 ± 2.6§ 16.8 ± 4.1§ 4.0~
11.5 ± 4.7§
10.4 ± 0.5*~ 5.0 ± 0.2a
10.6 ± 2.2*§ 13.5 ± 0.6*§ 12.0 ± 1.8§ 12.4 ± 0.6*§ 7.2 ± 1.4*§
100/100 13/35 38/54 19/58 23/79 26/45 16/49
7.0 ± 1.3*§ 9.2 ± 2.1H 16.8 ± 1.3*§
50/71 36/32
8.5 ± 2.1 *§
41/67
8.2 ± 2.7*§
#n=5; MST: mean survival time; *different from control (p< 0.05); chloroquine group (p< 0.05) using student t test, "two mice.
§
different from
304
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Table 3. Brine shrimp lethality and antimicrobial data for the extract and selected fractions from A. crassiflora
Compound
BSTb (llglmL)
ACWF01 27.7 ACWF02 3.8 ACWF03 2.9 ACWF05 4.6 ACWF06 1.6 ACWF030 1.8 ACW A Chloramphenicol a
b
Diametera (mm) of inhihition zone of bacterial growth
S. aureus
S. typhimurium S. pyogenes
E. coli
24h
48h
24h
48h
24h 48h
24h 48 h
NDc 7.0 ND ND 8.0 ND 12.0 20.0
ND 6.0 ND ND 7.0 ND 12.0 22.0
ND 9.0 11.0 10.0 11.0 8.0 14.0 22.0
ND 8.0 10.0 10.0 11.0 8.0 14.0 22.0
ND ND 11.0 11.0 15.0 9.0 14.0 20.0
ND 9.0 ND ND ND ND 10.0 ND
ND ND 11.0 11.0 14.0 9.0 13.0 19.0
ND 9.0 ND ND ND ND 10.0 ND
Diameter of inhibition zone minus diameter of the disc after 24 and 48 h observed for the tested extracts, fractions (100 Ilg/mL) and chloramphenicol (30 Ilg/disc). Brine Shrimp Lethality test. C Not detected.
ACKNOWLEDGEMENTS The authors thank F APEMIG for financial support (grant CDS-853/06), CNPq and PRPq for a scholarship to M.A.G. We also thank Dr. Jacqueline Aparecida Takahashi (Chemistry Department/UFMG) for arranging for the antimicrobial assays and Dr. Andreia M. Amaral (ICBIUFMG) for providing the bacteria strains. The technical support of Elaine A. Leite is also acknowledged. We also grateful to Albina Carvalho de Oliveira Nogueira for collecting the plant material.
REFERENCES Alali, F.Q., Liu, X.-X. and McLaughlin, J.L. (1999). Annonaceous acetogenins: recent progress. J. Nat. Prod., 62: 504-540. Bauer, A.W., Kirby, W.M.M., Sherriss, J.C. and Turck, M. (1966). Antibiotic susceptibility testing by a standardized single disk method. American Journal of Clinical Pathology, 45: 493-496. Bermejo, A., Figadere, B., Zafra-Polo, M-C., Barrachina, I., Estornell, E. and Cortes, D. (2005). Acetogenins from Annonaceae: recent progress in isolation, sythesis and mechanisms of action. Nat. Prod. Rep., 22: 269-303. Cave A., Cortes D., Figadere B., Laurens A. and Pettit G.R. (1997). In: "Progress in the Chemistry of Organic Natural Products", Springer-VerJag/Wien, Austria, pp. 81-288. Castro, I., Villegas, J.R., Soeder, B. and Castro, O. (1996). Oxoaporphine alkaloids from Annona purpurea wood. Fitoterapia, 67: 181-182. Fang, X-P., Rieser, M.J., Gu, Z-N., Zhao, G.-X. and McLaughlin, J.L. (1993). Annonaceous acetogenins: An updated review. Phytochem. Anal., 4: 49-67. Fisher, D.C.H., Gualda, N.C.A., Bachiega, D., Carvalho, C.S., Lupo, F.N., Bonotto, S.V., Alves, M.O. Yogi, A., Santi, S.M.D., Avila, P.E., Kirchgatter, K. and Moreno, P.R.H. (2004). In vitro screening for anti plasmodial activity of isoquinoline alkaloids from Brazilian plant species. Acta Tropica, 82: 261-266. Garavito, G., Rincon, J., Arteaga, L., Hata, Y., Bourdy, G., Gimenez A., Pinzon, R. and Deharo, E. (2006). Antimalarial activity of some Colombian medicinal plants. J. Ethnopharm., 107: 460-462.
Annona crassiflora Wood Constituents
305
Goulart, M.O.I., Sant'ana, A.E.G., Lima, R.A. and Cavalcante, S.H. (1993). Fitoconstituintes quimicos isolados de Jatropha ellhpt~ca. Atribui~ao dos deslocamentos quimicos dos atomos de carbono e hidrogenio dos triterpenos de jatrofolonas AeB. Quimica Nova, 16(2): 95-100. Guinaudeau, H. Leboeuf, M. and Cave, A. (1975). Aporphine alkaloids. (Lloydia) J. Nat. Prod., 38: 275. Hisham A., Pieters, L.A.C., Claeys, E.E., Dommisse, R. and Vlietinck, A.J. (1991). Squamocin-28-one and panalicin, two acetogenins form Uvaria narum. Phytochemistry, 30: 545-548. Hocquemiller, R., Cave A., Jacquemin, H., Touche, A. and Forgacs, P. (1982). Alcaloides des Annonacees, 36: alcaloides de l'Annona crassiflora Mart. Plantes Medicinales et Phytothtirapie, 16: 4-6, Hossain, M.S., Ferdous, A.J. and Hasan, C.M. (1995). Alkaloids from the stem bark of Desmos longiflorus. F~toterapia, 66: 463-464. Jossang, A., Dubaele, B. and Cave, A. (1990). Deux nouvelles acetogenins monotetrehydrofuranniques cytotoxiques: l'annomonicine et la montanacine. Tetrahedron Lett., 31: 1861-1864. Lima, L.A.R. dos S. (2007). Estudo Quimico e Avalia~ao Bio16gica das Sementes de Annona cornifolia A. St. -Hil. (Annonaceae) PhD Thesis, UFMG, Belo Horizonte, MG, pp. 317. Lorenzi, H. and Matos, F.J.A. (2002). Plantas medicinais no Brasil: nativas e ex6ticas cultivadas. Nova Odessa, SP: Instituto Plantarum. Peters, W., -Lin, L. Ze, Robinson, B.L. and Warhurst, D.C. (1986). The chemotherapy of rodent malaria, XL. Ann. Trop. Med. Parasitol., 80: 483-489. Pimenta, L.P.S. (1995). Estudo Quimico Bio-monitorado das sementes de Annona crassiflora objetivando 0 isolamento de acetogeninas tetra-hidrofuranicas. PhD Thesis, UFMG, Belo Horizonte, MG, pp. 268. Pimenta, L.P.S., Boaventura, Maria Amelia Diamantino, Sun, N.-J., Cassady, J.M. and Oliveira, A.B. (1996). Araticulin, a Bis-tetrahydrofuran polyketide from Annona crass~flora seeds. Phytochemistry, 42: 705-707. Pimenta, L.P.S., Prates, H.T., Viana, P.A. and Boaventura, M.A.D. (2000), Insecticide action of ethanolic extract from Annona crassiflora seeds against Spodoptera frugiperda In: XXI International Congress of Entomology, 2000, Foz do Iguassu - Parana. Abstract Book I, 1: 1035-1035. Pimenta, L.P.S., Nascimento, F.C., Assun~ao, A.C. and Boaventura, M.A.D. (2001). Laurifolin, a novel acetogenin from Rollinia laurifoha leaves. Tetrahedron Lett., 42: 8433-8434. Pimenta, L.P.S., Pinto, G.B., Takahasi, J.A., Silva, L.G.F. and Boaventura, M.A.D. (2003). Biological screenig of annonaceous Brazilian medicinal plants using Artemia salina (brine shrimp test). Phytomedicme, 10: 209-212. Pimenta, L.P.S., Lima, L.A.R. dos S. and Boaventura, M.A.D. (2008). Submitted for publication. Rupprecht, J.K., Hui, Y.H. and Mclaughlin, J.L. (1990). Annonaceous acetogenins: a review. J. Nat. Prod., 53: 237-278. Sahai, M., Singh, S., Sing, M., Gupta, Y.K., Akashi, S., Yuji, R., Hirayama, K., Asaki, H., Araya, H., Hara, N., Eguchi, T., Kakinuma, K. and Fuji Oto, Y. (1994). Annonaceous acetogenins from the seeds of Annona squamosa. Adjacent bis-tetrahydrofuranic acetogenins. Chem. Pharm. Bull., 42: 1163-1174. Santos, L.P., Boaventura, M.A.D. and Oliveira, A.B. (1994). Crassiflorina, uma acetogenina tetra-hidrofuranica citot6xica de annona crassiflora (araticum). Quimica Nova , 17: 387-391. Santos, L.A.R. dos, Pi menta, L.P.S., Boaventura, M.A.D. (2006). Cornifolin, a new bistetrahydrofuran annonaceous acetogenin from Annona cornifolia. Bwchemical Systematics and Ecology, 34: 78-82. Takahashi, J.A., Pereira, C.R., Pimenta, L.P.S., Boaventura, M.A.D. and Silva, L.G.F.E. (2006). Antibacterial activity of eight Brazilian Annonaceae plants. Nat. Prod. Rep., 20: 21-26. Zeng, L., Ye, Q., Oberlies, N.H., Shi, G., Gu, Z.-M., He K. and McLaughlin, J.L. (1996). Recent Advances in Annonaceous Acetogenins. Nat. Prod. Rep., 275-293.
"This page is Intentionally Left Blank"
16 Pharmacokinetics of Active Constituents of Rhodiola rosea SHR-5 Extract A. PANOSSIAN2,*, A. HOVHANNISYAN 1, H. ABRAHAMYANl,
E. GABRIELYAN 1 AND G. WIKMAN 2
ABSTRACT The active constituents of Rhodola rosea extract SHR-S of the popular plant adaptogen Rhodiola rosea are tyrosol, rhodioloside and rosavin. Validated capillary electrophoretic methods for the determination of these components in the blood was developed and used for a pharmacokinetic study in rats and human (n=16) volunteers. Rhodioloside was found to be quickly and completely absorbed into the blood of rats (bioavailability-7S-90%), distributed within organs and tissues, and rapidly metabolised to tyrosol following oral administration of SHR-S at doses of20 and SO mg / kg. Many of the measured pharmacokinetic parameters of rhodioloside were significantly different when the pure compound was administered rather than the extract. The basal level of tyro sol in blood plasma of rats increased following administration of SHR-S as a result both of absorption of free tyrosol present in the extract and of biotransformation ofrhodioloside into tyrosol, which occurred within the first 2 h. The pharmacokinetics and the rate ofbiotransformation ofrhodioloside were essentially the same following single or multiple regimes of administration of SHR-S. Rosavin has low bioavailability (20-26%) and was quickly eliminated from the blood of rats that have been administered SHR-S. The results of the study in humans showed that 16 healthy volunteers, after oral administration of two tablets ofRosenroot, reached a maximum concentration of rhodioloside and rosavin in blood plasma after 2 h, where the absorption rate constant was equal in 1. "ExLab" Expert Analytical Laboratory of Armenian Scientific Centre of Drug and
Medical Technology Expertise, Komitas Ave. 49/4, 375051 Yerevan, Armenia. 2. Swedish Herbal Institute, Prinsgatan 12, SE-413 05, Gothenburg, Sweden. * Corresponding author: E-mail: [email protected]
308
Camp. Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
both components. Although, in contrast to rhodioloside, rosavin was not detected in blood plasma at 0.5 hand 1 h after oral administration of two tablets ofRosenroot. The maximum concentration and elimination half-life ofrhodioloside was 2-3 x higher than in rosavin. The AUCo_oo Cmax and AUCO_t values of rhodioloside were 3.5, 2.2 and 3.1-fold higher than in rosavin. Thus, the elimination of rhodioloside from the blood was 1.8 fold longer than the elimination time ofrosavin. Key words : Capillary electrophoresis, pharmacokinetic, Rhodiola rosea L., rhodioloside, rosavin, tyrosol, SHR-5 INTRODUCTION
Rhodiola rosea is one ofthe most popular plant adaptogens in Russia. Since 1969 Rhodiola rosea has been recommended by the Pharmacological Committee of the Ministry of Health ofthe USSR for the medicinal use as a stimulant offatigue in patients experiencing aesthetic states or in healthy people with a tendency to asthenisation during their work, which requires high mental exertion or to recover their work capacity during and after long periods of intensive physical work. In addition, Rhodiola rosea is recommended in cases of borderline nervous-mental diseases, neuroses, neurosis-like disorders, and psychopathies. In psychiatric practice, Rhodiola rosea extract is indicated for the correction of neurological side effects of psychopharmacological therapy and for the intensification and stabilisation of remissions of aesthetic and apathico-abulic type schizophrenia patients (Saratikov et al., 1965; Krasik et al., 1970; Saratikov, 1973; Mikhailova, 1983; Brichenko et al., 1986; National Pharmacopoeia ofthe USSR, 1987; Saratikov & Krasnov 1987). A number of double-blind, placebo-controlled randomised clinical trials ofthe standardised extract (SHR-5) of R. rosea have shown that use of the drug can reduce general fatigue (as determined by tests measuring complex perceptive and cognitive cerebral functions) under certain stress conditions by increasing total mental performance (Darbinyan et al., 2000), and can significantly improve general well-being, physical fitness, mental fatigue, and coordination in healthy subjects when taken repeatedly for a short period of time (7-10 days) (Spasov et aI., 2000 a,b). A similar effect was observed after single dose administration of standardised SHR-5 extract on the capacity for mental work against a background offatigue and stress on a highly uniform population comprising of 161 cadets (Shevtsov et al., 2003). The anti-fatigue effect of Rhodiola SHR-5 extract, as well as improved attention in fatigue and stressful conditions, were earlier observed in patients with chronic stress induced fatigue (Olsson et al., 2008). Moreover, it has been beneficial in mild and moderate depression (Darbinyan et al., 2007).
Pharmacokinetics of Active Constituents of Rhodiola rosea
309
The active principles ofRhodiola extracts are tyro sol (syn. salidrosoD [2-( 4-hydroxyphenyDethanoll, and the phenolic glycosides, rhodioloside (syn. rhodosin, salidroside) [2-( 4-hydroxyphenyl)ethyl-l-~- D-glucopyranosidel and rosavin (syn. cinnamyl alcohol ~-vicianoside) [3-phenyl-2-propenyl-O-(a-Larabinopyranosyl-(1-6)-~-D-glucopyranosidel (Saratikov et al., 1968; Saratikov, 1973; Barnaulov et al., 1986; Sokolov et al., 1990; Linh et al., 2000; Panos sian et al., 2007; Panossian et al., 2008). In order to determine the contribution of individual components with respect to the overall curative effect of an herbal extract, it is necessary to study the pharmacokinetics of the active compounds in the particular preparation, since the presence of even inactive ingredients can influence the bioavailability, absorption, distribution, metabolism and excretion ofthe active compounds. The aim of the present study was to the investigate the pharmacokinetics of rhodioloside and rosavin in rats (Panossian et al., 2002) and healthy human volunteers after single and multiple oral administration of Rhodiola rosea SHR-5 extract-used as an active ingredient of ''Rosenroot ®" tablets used in Sc andinavia as an adaptogen to increase impaired performance in fatigue and weakness.
MATERIALS AND METHODS Chemicals HPLC-grade methanol (RotisolV®; # 7342.1) and acetone (# 7328.2) were purchased from Carl Roth (Barcelona, Spain); HPCE-grade water (# 506285780), 50 mm borate buffer (pH 9.3; # 5062-8573) and O.IM sodium hydroxide (# 5062-8575) were from Hewlett-Packard (Palo Alto, CA, USA); ethyl acetate (# 15485-7; lot KI 01058DI) and ~-glucuronidase (# G-7770; lot 22K8942) were from Sigma-Aldrich (St Louis, MO, USA); and heparin lock flush solution (containing 500 USP heparin units/mL) was from Abbott Laboratories (Chicago, IL, USA). Unless stated otherwise, the water employed was deionised and distilled.
Study drug Rhodiola rosea L. roots soft extract SHR-5 (batch No EX 20465, Swedish Herbal Institute, Sweden), quantified for rhodioloside content, (10 ± 2 mg/g), rosavin (20 ± 3 mg/g) and tyrosol (1.5 ± 0.3 mg/g). Rosenroot tablets (400 mg, batch Nr L211) contain 144 mg SHR-5, 3.63 mg rhodioloside and 4.2 mg rosavin.
Standard Solutions Reference standards stock solution. T A standard solution of the reference compounds tyrosol (Swedish Herbal Institute, Goteborg, Sweden; series 22.10.2001), rhodioloside (VILAR, Moscow, Russia; series S10402) and rosavin (VILAR; series S10402) was prepared by accurately weighing 5.0 mg of each
310
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
compound into a 5 mL volumetric flask, dissolving and adjusting to volume with methanol
Study animals Three hundred time-mated male Wistar rats were obtained from the Institute of Fine Organic Chemistry of the National Academy of Science, Yerevan, Armenia. All animals were clinically examined upon arrival and any animal showing signs of abnormality or disease was replaced before the start of the study. No animals were replaced after the study had commenced. Animals were kept for 10-15 days prior to the start ofthe study under a 12112 h light/ dark cycle and had free access to standard rat chow. During the study, a standardised diet for rats (CombilYerevan, Combi-Corm plant; for composition, Zapadnyuk et al., 1983) was provided, but feeding was discontinued before administration of a test substance. Only tap water was offered ad lib. Throughout the observation period, animals were kept separately in cages (55 x 35 x 25 cm), each consisting of a polystyrene case and a lattice-framed steel lid, and supplied with wood-sawdust bedding. A 12112 h light/dark cycle was maintained in the animal room throughout the study period, and the temperature and humidity were 22 ± 4°C and 40 ± 5%, respectively, with 1-2 changes ofairlh. Each day, or after completion of work, the floor of the animal room was swept and mopped with an atmospheric biocide-cleanser. Excretions were collected every second day on trays lined with absorbent paper which were suspended beneath each cage.
Dosage and Administration Dosage, Sampling, Drug Safety Control. The concentrations of rhodioloside, rosavin and tyrosol in the SHR-5 solutions were analysed by HPCE before their administration to the animals. The administered concentrations were 57.6, 97.5 and 6.5 flg/mL, respectively. In addition, two doses ofSHR-5 were used in this study. The 20 mg/kg dose shows maximum antiarrhythmic and antistressor effect in rats (Saratikov, 1973; Sokolov et al., 1990) and its rhodioloside content was equivalent to a human therapeutic dose (10 drops of Rhodiola extract), which is close to one tablet of "Rosenroot ®". The 50 mg/kg dose contained rhodioloside equivalent to a human mean daily dose (25 drops of Rhodiola extract) (Saratikov, 1973), which corresponds to two tablets of"Rosenroot®". Three sets of experiments aimed at determining the pharmacokinetics of tyrosol, rhodioloside and rosavin in the blood of animals were performed, whilst two further groups of animals were used to study the biotransformation of the active components of SHR-5 (Table 1).
Pharmacokinetics of Active Constituents of Rhodiola rosea
311
Table 1. Experimental design ofthe study of pharmacokinetics and biotransformations following administration of special extract SHR-5 of Rhodiola rosea L. to rats
....5 ....~ til
=
~ 1
SHR-5 54
20
Single
I.v.
2
2
SHR-5 54
20
Single
Oral
10
3
SHR-5 54
50
Single
OralIO
4
SHR-5 48 20/daily Multiple (5 days) 5 SHR-5 36 50/daily Multiple (5 days) Single 6 Rhodiolo-42 0.570 side 7 Water 42 Single
Oral
10
Oral
10
Oral
10
Oral
10
0.026 0.230 0.390 0; 0.06; 0.33; 0.5; 0.75; 1.0;1.5; 2.0; 3.0 0.026 0.230 0.390 0; 0.5; 1.0;1.25; 1.5; 2.0; 4.0; 5.0; 6.0 0.065 0.576 0.975 0; 0.5; 1.0; 1.25; 1.5; 2.0; 4.0; 6.0; 8.0 0.026 0.230 0.390 0; 0.5; 1.0; 1.25; 1.5; 2.0; 4.0; 6.0; 0.065 0.576 0.975 0; 1.0; 1.5; 2.0; 4.0; 5.0; 0.576 0; 0.5; 1.0; 1.5; 2.0; 3.0; 5.0 0; 0.5; 1.0; 1.5; 2.0; 3.0; 5.0
Pharmacokinetic Studying Rats In the first set ofpharmacokinetic experiments, SHR-5 was administrated intravenously via the tail vein at a dose of20 mg/kg to 48 animals in group 1 (mean weight 112 ± 10 g; range 100-120 g). Blood samples (4-6 mL each) were collected in heparinised centrifuge tubes immediately before administration of drug and at various times between 0.06 and 3 h after injection (Table 1). Following collection, blood samples were centrifuged at 300 x g for 15 min to obtain blood plasma which was stored at -20°C until required for analysis. In the second set of experiments, SHR-5 was administrated orally to two groups of 48 animals that had been deprived of food for 12 h: free access to food was provided 2.5 h after the administration of the drug. Animals in group 2 (mean weight 167 ± 10 g; range 150-180 g) received SHR-5 solution at a dose of20 mg/kg, whilst those in group 3 (mean weight 120 ± 8 g; range 110-130 g) received a dose of 50 mg/kg. Blood samples (4-6 mL each) were collected in heparinised centrifuge tubes before administration and at various times between 0.5 and 6 h after administration (Table 1). In the third set of experiments, oral administration of SHR-5 was repeated for 5 days at daily doses of 20 mg/kg (group 4: mean weight 157
312
Camp. Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
± 8 g; range 150-180 g) and 50 mg/kg (group 5: mean weight 145 ± 10 g; range 130-160 g). Animals were deprived of food for 12 h prior to the administration (at 9.00 pm) of a single daily dose of SHR-5 solution: 2.5 h after drug administration, animals were allowed free access to food until 9.00 am. Blood samples (4-6 mL each) were collected in heparinised centrifuge tubes before administration and at various times between 1.0 and 5 h after the fifth administration (Table 1).
Study ofBiotransformation ofRhodioloside In order to study the biotransformation of active compounds, a group of 36 animals (group 6; mean weight 125 ± 9 g; range 110-140 g) was fasted for 16 hand rhodioloside was administrated orally at a dose of 0.570 mg/kg. Mter administration of the drug, only water was supplied during the whole experiment. Blood samples (4-6 mL each) were collected in heparinised centrifuge tubes before administration and at various times between 0.5 and 5 h after administration (Table 1).
Human Study Design The study was performed as a blind randomized study, in compliance with the revised declaration of Helsinki (Hong Kong, 1989) and The Ministry of Health in Armenia.
Volunteers The clinical part of the study was carried out at the Centre of Traumatology, Orthopedics, Burns and Radiology at the Ministry of Health of Armenia and written informed consent was obtained from each subject before the study was initiated. In total, 16 normal and healthy volunteers were scheduled to participate (two groups of8-male and 8-female). All volunteers had been donors at the Centre of Hematology for more than four years and a written and signed agreement between the head of investigation and the volunteer were included in the study. Prior to the study, each subject underwent a brief physical examination and had blood drawn for serum chemistry and hematology analysis, according to the inclusion! exclusion criteria. During the study each volunteer received a number and a special code, in compliance with their sex.
Study Schedule The study included the following stages: selection of volunteers, receiving written signed agreements (information for volunteers), medical examination and blood biochemistry and hematology analysis, disposition in groups, drug administration and blood sampling, determination of drug in blood samples, and calculation of pharmacokinetic parameters. The study was provided according to the scheme notified in the investigation protocol.
Pharmacokinetics of Active Constituents of Rhodiola rosea
313
Selection ofVolunteers The volunteer inclusion criteria focussed on healthy males and females aged 28-55, without allergenic anamnesis or illnesses, such as chronic diseases of the kidney, liver, cardiovascular and nervous system. In addition, the volunteers have to have signed an agreement written by the head of the investigation. Finally, they needed to be able to collaborate adequately during the entire length of the study. The volunteer exclusion criteria concerned volunteers suffering from chronic illnesses, such as cardiovascular diseases, diabetes (etc.), and arterial hypotension (etc.). In addition, volunteers addicted to, for example, medicines, narcotics and tobacco were also excluded. Finally, they were not allowed to participate in any other trials. In addition, volunteers were removed from the investigation ifthey experienced an individual reaction, adverse reactions, serious adverse events or if their health and physical condition deteriorated.
The Dates ofVolunteers Enrolled in the Study Bearing in mind the expected intra-individual variability, 16 healthy volunteers (Caucasians), which had not received any drugs in the past month prior to this study were enrolled. The volunteers, aged 28-55, were both female (mean age 41.37 ± 11.6) and male (mean age 45.50 ± 6.9).
Dosage, Blood Sampling and Safety Control Dosage and Blood Sampling The subjects were fasting during one night and then received two tablets of Rosenroot with 200 mL water two h before breakfast at 9 a.m. The standard breakfast time was at 9.30 a.m. -10.00 a.m., while the standard dinner time was 15.30-16.00. The period measured was 10 h. In addition, the subjects agreed to refrain from the use of other prescription or nonprescription drugs (including vitamins), alcohol and coffee during the entire study period. At the end of the study, each subject underwent a brief physical examination and had blood drawn for serum chemistry and hematology analysis. The volunteers were all Armenian and live in the capital Yerevan. A 20-gauge heparinised catheter was inserted into a vein of the forearm to collect blood samples in a dilute heparin solution (10 to 15 VI mL). Two millilitres ofblood was drawn and discarded prior to collection of each blood sample (6-8 mL) into heparinised tubes. These tubes were centrifuged at 300 g for 15 min to prepare plasma and stored at _20DC until further analysis. For each volunteer, a set of blood samples (6-8 mL each) were drawn before (0) administration and at 0.5, 1.0, 1.5,2.0,3.0,4.0,6.0, 8.0 and 10.0 h thereafter.
314
Camp. Bio. Nut. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
Safety Control Drug safety was rated from physical examination, measurement of blood pressure, heart rate, and laboratory examination, such as hematology (leukocytes, erythrocytes, and hemoglobin), biochemistry and documentation of adverse events (AE), which were asked and recorded by the clinical staff every h after drug intake. The classification of the AE's was in accordance with the EC-guideline 111/3445/91-EN and termed as mild, moderate and severe. In addition, the causes were classified as definite, probable, possible, unlikely, not related and not possible to judge.
HPCE Analyses In order to analyse rhodioloside and rosavin in blood, 1.5 mL of acetone was added to 1.5 mL of the plasma sample, the mixture was vortex-mixed, a further 5 mL of acetone added, the whole vortex-mixed again and the proteins precipitated at 4°C by centrifugation at 2000 x g for 15 min. The supernatant was removed, evaporated to dryness at 40°C using a vacuum rotary evaporator and the residue dissolved in 100 mL of methanol, transferred into HPCE vials and subjected to analysis. For the analysis of tyrosol in blood, 1.0 mL of methanol was added to 1.0 mL of the plasma sample, the mixture was vortex-mixed, a further 3.0 mL of methanol added, the whole vortex-mixed and the proteins precipitated at 4°C by centrifugation at 2000 x g for 15 min. The supernatant was removed and treated exactly as described above. For the analyse of tyro sol in urine, ca. 50 J..ll of p-glucuronidase solution was added to 1.0 mL of urine sample, the whole mixed vigorously for 30 s and then incubated at 55°C for 3 h. After incubation, the mixture was purified via solid phase extraction by applying the sample to Supelclean LC-18 SPE tubes (Supelco, Belefonte, PA, USA; 3 mL; lot SP1662A), which had been pre-washed with 1 mL of methanol and 1 mL of water, and eluting with 3 mL each of water : methanol mixtures of compositions 95:5, 85:15 and 50:50. The second and third eluents were evaporated to dryness at 50°C using a vacuum rotary evaporator: the residue was dissolved in 1.0 mL of acetate buffer (pH 5.2), 5.0 mL of ethyl acetate added, the whole vortex-mixed for 60 s, and the two layers separated. The organic phase was evaporated to dryness at 50°C using a vacuum rotary evaporator, the residue was dissolved in 100 ml of methanol and subjected immediately to HPCE analysis. Analyses were carried out using a Hewlett Packard HPCE system comprising a model HP3DCE apparatus interfaced to a HP Kayak XA workstation and a HP Laser Jet 4000 printer, and equipped with a HP fused silica capillary (# G 1600-61232; total length 56 cm; effective length 50 cm; i.d. 50 mm; optical path length 150 mm) maintained at lOoC. The column was preconditioned by flushing for 1.0 min. with 0.1 M sodium hydroxide, rinsing with water, and then flushing with mobile phase [50 mM
-. •
•
• •
. !II
•
.. +
:II
I)
• •
1
t2.
1
13..
Rhodiol Midc-
1
t".
Fig 1. Electropherogram of standard solutions of salidroside and rosavin (25 mg/l) monitored at 201 nm [the solvent system was 50 mM borate buffer (pH 9.3): methanol (85:15, v/v): the migration times for salidroside and rosavin were 11.86 ± 0.06 and 12.39 ± 0.06 min, respectively: the relative migration time salidroside/rosavin was 0.9573 (CV = 0.09%))
mt,
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
316
borate buffer (pH 9.3) : methanol in the ratio 85:15 (v/v) for rhodioloside and rosavin, and 60:40 (v/v) for tyrosol] for 2 min. Injection was at 50 mbar for 4 s, the running voltage was 25 kV, detection was at 201 nm (reference 300 nm), 251 nm (reference 450 nm) and 275 nm (reference 400 nm), and the run time was 15 min. The capillary was washed for 5 min. after very fifth run and the mobile phase replaced. Fig 1 shows a capillary electropherogram ofrhodioloside and rosavin (migration times 11.86 ± 0.06 and 12.39 ± 0.06 min, respectively), whilst Fig 2 depicts the electropherogram for tyrosol (migration time 12.79 ± 0.10 min). Calibration curves for all analytes were linear in the range 2.50 125.00 mgll with correlation coefficients of 0.9986, 0.9962 and 0.9984, respectively, for tyrosol, rhodioloside and rosavin. The limit of detection for all analytes was 1.0 mgll (at a signal! noise ratio of 3) and the limit of quantification was ca. 2.5 mgll. The accuracy was 98.59% for rhodioloside, 97.20% for rosavin and 97.34% for tyrosol. The recoveries were 90.38 and 93.82%, respectively, for rhodioloside and rosavin from blood samples, and 86.5 and 99.5%, respectively, for tyrosol from blood and urine samples. The repeatability for all analytes was ca. 94%, and the relative standard deviation (RSD) value was established at <5% for all validation parameters.
m
4
o
- -."'-- . "..-.....-~ . _.,
.-
\ I
I
I
\.J.I m
Fig 2. Electropherogram and UV spectrum of a standard solution of tyrosol (25 mg/l) monitored at 201 nM [the solvent system was 50 mm borate buffer (pH 9.3): methanol (60:40, v/v): the migration time for tyrosol was 12.615 min.)
Calculation of Concentration of Analytes in Blood and Urine Samples The mean concentrations ofrhodioloside and rosavin in blood plasma (CMEAN were calculated from the mean values measured by HPCE (CMEAN HPCE) using the equation CMEAN PLASMA =CMEAN HPCE / (K1 X ~), where Kl is the concentration factor of the sample during pre-treatment (= 15) and ~ PLASMA)
Pharmacokinetics of Active Constituents of Rhodiola rosea
317
is the appropriate coefficient of recovery (0.904 and 0.938 for rhodioloside and rosavin, respectively). The mean endogenous concentration of tyro sol (C MEAN ENDOGENOUS) in blood plasma and urine was determined from CMEAN ENDOGENOUS =CMEAN HPcE"(Kl X ~), where Kl is the concentration factor of the sample during pre-treatment (= 10) and ~ is the appropriate coefficient of recovery (0.869 and 0.995 for blood plasma and urine, respectively). The mean exogenous concentration of tyrosol in blood plasma after administration of drug (CMEANPLASMA) was calculated from CMEANPLASMA = CMEAN TOTAL - CMEAN ENDOGENOUS' where CMEAN TOTAL is the mean total concentration (endogenous + exogenous) oftyrosol found by HPCE in blood plasma after administration of a drug, and CMEAN ENDOGENOUS' is the mean endogenous concentration of tyro sol in control samples of blood plasma. The amount of rhodioloside excreted in the urine as tyrosol and expressed as a percentage of the amount of SHR-5 administered (DE)' was determined from: DE =
(R2 - R 1) x t (k x Ds + D FT ) x BW
x V x 100 t
where Rl is the rate (mg/mUh) of urinary excretion of endogenous tyro sol before administration of SHR-5, R2 is the rate (mg/mLlh) of urinary excretion of tyro sol after administration of SHR-5, t is the time (h) after administration of SHR-5, k is the molecular weight ratio of tyrosol : rhodioloside (= 0.46), Ds is the dose (mglkg) ofrhodioloside administrated, DFT is the dose (mglkg) offree tyro sol administrated, BW is the total body weight (kg) ofthe two animals in the chamber, and Vt is the total volume (mL) of urine collected over time t.
The Measured Pharmacokinetic Parameters The pharmacokinetic parameters were calculated using the TOPFIT software (version 1.1; Godecke, Freiburg, Germany; Schering, Berlin, Germany, Thomae, Biberach-an-den-Riss, Germany): parameters for multiple administrations were calculated using the method ofDost (1968). The measured parameters were: (i) CMAX (ng/mL) - the maximum concentration taken directly from the concentration course; (ii) Co ng/mL initial drug concentration in blood plasma after the first dose; (iii) F (%)bioavailability; (iv) KA (Ih) - the absorption rate constant; (v) KEL (Ih) - the elimination rate constant calculated from a logllinear regression of the concentration/time data (terminal slope); (vi) tV2 (h) - the elimination halflife (O.693lkEL ); (vii) AUC o_oo (f.,lg.h/mL) - area under the curve after extrapolation from time x to infinity, where x is the last time point with a concentration above the lower limit of quantification (= AUCO-t + CxfKEL); (viii) CIt (mUmin) - clearance (= F x doseiAUC o. ,); (ix) MRT (h) - mean residence time (= AUMC IAUC where AUMC is the area under the first statistical moment curve - Io. ¥ t.C.dt (ng/mL)h2); (x) K12 (Ih) - distribution
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
318
rate constant for the transfer of drug from central to peripheral compartment; (xi) ~1 (Ih) - distribution rate constant for the transfer of drug from peripheral to central compartment; (xii) Vc (1) - apparent volume of distribution of central or plasma compartment (= D/Co); (xiii) Vnss (l) - apparent volume of distribution at steady state (= F x dose/Co or K12 + ~1~1 x Vc for a one or two-compartment model, respectively); (xiv) CssMAX(nglmL) - maximum drug concentration in blood plasma in multiple dosing at steady state; (xv) CSsAV (ng/mL) - mean steady state blood plasma concentration; (xvi) CSSMIN(ngl mL) - minimum drug concentration in blood plasma in multiple dosing at steady state; and (xvii) 't (h) - dosing interval.
RESULTS Pharmacokinetics Study in Rats Rhodioloside Following intravenous injection of SHR-5 at a dose of 20 mg/kg, the concentration ofrhodioloside in blood plasma decreased rapidly from 920 to 367 ng/mL after 30 min., and then diminished slowly reaching a value of 190 ng/mL after 1 h (Fig 3). When SHR-5 was given orally at a dose of 20 or 50 mg/kg, the maximum concentration of rhodioloside in plasma was attained 1 h after administration (Fig 3). For both doses, the concentration ofrhodioloside fell sharply during the interval from 1-2 h after administration, and then declined slowly. Mter 1 h following intravenous injection of SHR-5, or 5-6 h after oral administration of the drug, rhodioloside was no longer detectable in blood plasma since the level of the analyte fell below the limit of detection (100 ng/mL) of the analytical method.
-g>
E
c::
o
~
1:
go
U
1---------------- limit of detection 2
3
4
5
6
Time (h) Fig 3. Profiles of plasma concentration of salidroside versus time following a single intravenous (20 mg/kg, -. -) and oral administration of SHR-5 solution at doses of 20 mg/kg (d) or 50 mg/kg (0). The values are means of replicate (n = 6) measurements with their respective standard deviations
Pharmacokinetics of Active Constituents of Rhodiola rosea
319
Fig 4 shows that after the fifth oral administration of SHR-5 at a dose of 50 mg/kg, the maximum concentration of rhodioloside in plasma was reached 1-1.5 h after drug administration. During the period 1-2 h after administration, the concentration of the analyte decreased sharply, following which the decline was much slower. Around 5 h after multiple administration of SHR-5, the blood level of rhodioloside fell below the limit of detection.
~
E 750
0,
~ !\
c c 0
..
;:I
500
E u
C
0 U
~
250
~
0
0
.~
it:. ' I
14 I
~
I
~--- L
~ ,
24
I
I
I
,
~
I
I~
G)
~
6.
48
'-
t
72
--- ~
"-
1 1 i 1 i 1th
2th
3
th
4th
120
96
5th
H admmistrations
Fig 4. Profiles of plasma concentration ofrhodioloside versus time following multiple oral administration of SHR-5 solution (50 mg/kg/day) showing experimental values (0) and calculated values (M based on single dosing; each are means of replicate (n = 6) measurements with their respective standard deviations
The pharmacokinetic parameters for rhodioloside, determined for all routes of administration and doses used in the study, are shown in Table 2. Clearly, rhodioloside was rapidly and completely absorbed into the blood (KA - 0.7-2.0 /h; F - 78-93%) after single and multiple oral administrations of SHR-5 to rats at doses of 20 and 50 mg/kg. Following oral administration, rhodioloside was quickly distributed within tissues but returned somewhat slowly to the circulatory blood system (Kl~l > 2). For both dose levels studied, no significant differences were found in the values of the pharmacokinetic parameters of rhodioloside following single and multiple oral administrations of the drug. The rates of absorption, distribution and elimination after administration of SHR-5 at a dose of 20 mg/kgldaily were, however, lower than those following administration of the drug at a dose of 50 mg/kgldaily, but the bioavailability for both doses was the same (Table 2). Analysis of the pharmacokinetic curve of rhodioloside plotted with semi-logarithmic coordinates showed that in all cases the pharmacokinetics could be described by a two compartment
320
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
pharmacokinetic model. The differences between the concentrations of rhodioloside in plasma measured experimentally and those calculated using pharmacokinetic parameters for single dose administration, were not significant. Table 2. The comparison of pharmacokinetic parameters of rhodioloside after SHR-5 administration Route of administration
Parameters I.V. single Doses,
20
Oral single
Oral multiple after Sih administration
20
50
20
50
2.06 ± 0.06 400 ± 8
0.95 ± 0.07 700 ± 26
0.73 ± 0.13 472 ± 9
1.70 ± 0.18 545 ± 8
0.023 ± 0.001 1121 ± 27
0.025 ± 0.006 1703 ± 33
0.040 ± 0.002 1013 ± 23
0.059 ± 0.012 2114 ± 61
4.781 ± 0.770
1.446 ± 0.267
0.582 ± 0.200
1.301 ±0.033
0.720 ± 0.020 0.560 ± 0.060
0.392 ± 0.088 0.531 ± 0.011
0.185 ± 0.024 0.350 ± 0.008
0.661 ± 0.16 0.536 ± 0.02
3.57 ± 0.18
4.45 ± 0.64
5.59 ± 0.52
3.48 ± 0.35
mg/kg
Ka, h·l 974 ± 27 C max, ng/mL 0.087 ± 0.020 Vdss, 1 AUCO-<.o 809 ± 26 [(ng /mL) 'hl K 1I2, h· l 0.738 ± 0.316 Km h·l 1.493 ± 0.265 CIt' mLlmin 1.681 ± 0.084 t 1l2, h
0.57 ± 0.04
Rosavin Following intravenous injection of SHR-5 at a dose of 20 mg/kg, the concentration of rosavin in blood plasma fell rapidly from 775 to 274 ng! mL after 30 min, and then diminished slowly (Fig 5). The maximum plasma concentration ofrosavin following single oral administrations ofSHR-5 at doses of 20 and 50 mg/kg was attained 1 h after drug administration, following which the blood level of the analyte sharply decreased within 11.5 h and then declined slowly to a level (<100 ng!mL) which was no longer detectable (Fig 5). It is noteworthy that the HPCE method developed for the determination ofrosavin in blood plasma only allowed detection of the analyte between 1-1.5 h after oral administration of SHR-5 at a dose equivalent to a single therapeutic human dose of the drug: in all other blood samples the concentration of rosavin was below the limit of detection of the method. This may be explained by the very low bioavailability of rosavin (F =20-26%; Table 3) and its rapid disappearance (t lf2 =0.5-0.6 h). Similar results were found following multiple administration of SHR-5 at a dose of 50 mg/kg one time daily where, again, it was possible to determine the analyte in blood only between 1-1.5 h following the fifth administration. The results demonstrate that the concentration of rosavin determined 1 h after the fifth administration ofthe drug (620.2 ± 131.0 ng!mL) is somewhat variable and near to the calculated value ofCSSMAX (574 ng/L).
321
Pharmacokinetics of Active Constituents of Rhodiola rosea
1000
E
0, 800 c c 600 ~
0
.~
....
C 400 ~ c 0
U
200 0 0.0
0.5
1.5
1.0 Time (h)
2.0
Fig 5. Profiles of plasma concentration of rosavin versus time following a single intravenous (20 mg/kg, -. -) and oral administration of SHR-5 solution at doses of 20 mg/kg (V ) or 50 (0 ) mg/kg and The values are means of replicate (n=6 ) measurements with their respective standard deviations Table 3. The comparison of pharmacokinetics parameters of rosavin after SHR-5 intravenous and oral administration Route of administration
Parameters
LV. single
Oral single
20
20
Doses, mg/kg Ka, h- I C max, nglmL Vdss, I AUCo.,,)( ng ImL ) ·hj Clt' mUmin t l12 , h
F ,%
525.3 0.054 485 1.30 0.532
± ± ± ± ±
15.1 0.007 56 0.11 0.065
100
0.489 249.6 0.058 171 1.37 0.469
± ± ± ± ± ±
20.3 ±
Oral single 50
0.017 19.4 0.008 10 0.08 0.026 1.9
1.935 ± 579.1 ± 0.057 ± 492 ± 1.76 ± 0.631 ±
0.438 30.2 0.001 73 0.12 0.044
26.0 ± 6.5
Tyro sol It is a well-known fact that tyrosol is consumed through food and remains at a stable level in the blood and urine during fasting conditions (Visioli, 2000: Miro-Casas et at., 2001). Preliminary experiments showed that the basal level of free tyrosol in the blood plasma of rats subjected to a wash out period was 499.8 ± 15 ng/mL. After intravenous administration of SHR-5 at a dose of 20 mg/kg, the concentration of exogenous tyrosol in the plasma increased, reaching a maximum value after 1 h , but within 3 h the level decreased sharply and returned to the endogenous basal level (Fig 6). Following oral administration of SHR-5 at dose s of 20 and 50 mg/kg, the concentration of exogenous tyrosol attained its maximum value within 1.5 and 2.0 h , respectively, and then decreased exponentially (Fig 6 ).
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical E valuation I
322 . - 800
.....E0> C
'-'
700
c 0
600 ~ '-
-8 c:
c 500
- - Endogenous wmso1
0
0
400 I
I
I
I
0
2
3
4
5
6
Tir1e (hours) Fig 6. The mean plasma concentration vs time profiles for tyrosol after a single intravenous (20 mg/kg, _e -) and oral administration of SHR-5 solution of two doses: 20 mglkg ( -\7-) and 50 mg/kg-o-). The values are mean ± SD, n=6
In order to establish whether tyrosol could be generated from rhodioloside in vivo, the pharmacokinetic oftyrosol and rhodioloside were studied following oral administration ofrhodioloside at a dose of 0.570 mg/ kg (corresponding to that amount of rhodioloside present in a 50 mg/kg dose ofSHR-5). The concentration/time courses of tyro sol and ofrhodioloside in the blood plasma of rats fed with rhodioloside are shown in Fig 7. It was observed that the concentration of tyro sol reached its maximum value within 1 h, fell sharply within the period 1-1.5 h after administration, and then followed an exponential decrease. Within the first 2 h following administration ofrhodioloside, the concentration of tyro sol was significantly higher than that of rhodioloside, but after 2 h their concentrations were very similar. These results can only be explained by assuming that tyrosol measured in vivo was derived (enzymatically or as a result of hydrolysis in acidic conditions in stomach) from the rhodioloside administered to the animal, scheme 1. The comparison ofpharmacokinetic curves and parameters (Fig 7 & Table 4) ofrhodioloside after administration ofSHR-5 or rhodioloside showed that its concentration in blood plasma was significantly higher, particularly in the first 1.5 h after administration. The results also showed that the maximum concentration time ofrhodioloside in blood plasma appeared later when rhodioloside was administered alone, as a pure substance, in contrast to SHR-5 administration. Some other pharmacokinetic parameters were also significantly different after rhodioloside administration (as a pure substance), where the values ofC max , AUC o_oo ' tl/2 were less than 1.2 x - 2.0 x., in comparison to rhodioloside administration in SHR-5. The half-life time ofrhodioloside was shorter and the total amount in the blood was less
Pharmacokinetics of Active Constituents of Rhodiola rosea
2-( 4-Hydroxyphenyllethano I-I-b- Dglucopyranoside (rhodiolosidel
323
2-( 4-Hydroxyphenyl lethanol (tyrosoll
Schemel
~
E
18:)01600
-
q
~
Tyrosol
c
Rhodioloside
•
Tyrosol from SHR-5
•
Rhodioloside from SHR-5
.~.
~ 1400-
'C'1200 .Q 1000-
o
BasalEvelofendogenous~sol
.'
.
'
:
F~ 600 800-~ . ¥ - --+--. '~ . _ 0 . -. -- .. - -'
.3 ~::~~_~._~_:._ :::"-::::=_ _~=' o~ o
1
:-= ___
i
:
~
3
.. ;Q
i
-
4
5
Time (h) Fig 7. The mean plasma concentration vs time profiles for tyrosol (-0-) and rhodioloside (-0-) in the dose of 0.570 mg/kg after single oral administration ofrhodioloside, and tyrosol (_ e _) and rhodioloside (_ e _) after single oral administration of SHR-5 solution in doses 50 mg/kg, n=6. The values are mean ± SD
when rhodioloside was administered alone. At the same time, its distribution rate in tissues was almost the same since the values of the volumes of distribution were similar when both pure rhodioloside and SHR-5 were administered. Thus, the differences observed might be due to the presence of other chemically similar compounds in SHR-5, which might have been metabolised by the same enzyme as rhodioloside. Ifthis suggestion is correct, it means that the tyro sol content in blood plasma is higher after administration of pure rhodioloside than after administration ofSHR-5. The comparison of pharmacokinetic curves of tyrosol after administration ofSHR-5 and rhodioloside showed that after administration ofrhodioloside its concentration in blood plasma was significantly higher, particularly in the first 1.5 h after administration (Fig 9). In the interval between 2-3 h after administration, the concentration of tyro sol decreased approximately at the same rate, while at 3 h it reaches the basal level in both cases. Moreover, the maximum concentration of tyro sol was detected only 1 h after intravenous administration of SHR-5. This fact may also
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
324
indicate that the increase in tyrosollevel detected in the blood originated not only from free tyrosol of SHR-5, but also from biotransformation of rhodioloside and/or other precursors present in SHR-5. Table 4. The comparison of main pharmacokinetic parameters of rhodioloside in rats after oral administration ofrhodioloside and SHR-5 (rhodioloside dose: 0.570 mg/kg). The difference is significant at p<0.05. The values are mean ± SD, (n=6) Rhodioloside afterSHR-5 administration
Parameters
Rhodioloside after rhodioloside administration
t max , h Cmax ' nglmL
1,44 ± 0,18 334 ± 7 0.127 ± 0.011 1370 ± 46 0.846 ± 0.026
1,01 700 0.155 1702 0.682
±
2.58 ± 0.39
4.54
Vdss ' I AUC o", ' [(ng ImL) ehl Clt'mUmin t1f2,
h
Significance of the dereference, p value
±
0,03 26 0.007 40 0.016
0.0013 <0.0001 0.0660 0.0003 0.0004
±
0.40
0.0055
± ± ±
Pharmacokinetics ofRhodioloside and Rosavin in Human The study showed that the maximum concentration ofrhodioloside, in blood plasma, was reached 2 h after oral administration of two tablets of Rosenroot. Although, in the interval between 1 h - 8 h the concentration decreased exponentially, while after 8 h the level of rhodioloside was not detection (concentrations in blood plasma was less then the limit of detection) (Fig 8). In contrast to rhodioloside, rosavin was not detected at 0.5 and 1 h in blood plasma after oral administration. The maximum concentration of rosavin in blood plasma was reached after 2 hand decreased exponentially in the interval between 1-8 h after administration the concentration (Fig 8).
E
1000
0,
.s c
750
~
500
o
C
Q)
g o
250
o
2
345 Time (h)
6
7
8
Fig 8. The mean plasma concentration vs time profiles for mean concentration of rhodioloside (-0-) and rosavin (-e-) in blood plasma of volunteers after a single oral dose of two tablets of Rosenroot. The values are mean ± SD, n=16
325
Pharmacokinetics of Active Constituents of Rhodiola rosea
The study showed that the absorption rate constant and t max of rhodioloside were similar to that ofrosavin (Table 5), while the values of Cmax ' AUC O_t and AUC o_",' of rhodioloside were significantly greater (2.12x, 3.14x and 3.7x, respectively), in comparison to rosavin. Therefore, the elimination ofrhodioloside from the blood took longer (1.8x) than rosavin. Table 5. The comparison ofpharmacokinetic parameters ofrhodioloside and rosavin in human volunteers evaluated through individual parameters. The mean ± SD, n=16. The difference is significant at p<0.05 Parameters
Dose, p.g Ka,h- l Cmaxng/mL tmax,h AVCO_t , ng ·h/mL AVC o"" ' p.g .h/L t l12 , h AUCojDose
Rhodioloside
Rosavin
Ratio
A
B
A:B
9340 1.06 ± 0.19 948 ± 112 2.0 2569 ± 344 4477 ± 645 2.5 ± 0.7
7740 1.04 ± 0.13 446 ± 90 2.0 819 ± 142 1192 ± 169 1.4 ± 0.3
1.2 1.0 2.12 1.0 3.14 3.7 1.8
0.48
0.26
1.8
Significance of the dereference, p value
>0.05 <0.001 >0.05 <0.001 <0.001 <0.001
Table 6. Comparison of the main pharmacokinetic parameters of tyro sol, following oral administration of salidroside and SHR-5 (both in a dose containing 0.570 mg/kg salidrosidel to rats, as calculated from the mean concentrations of exogenous tyrosol in the blood Parameters
KA (/hl tMAJ{ (h) CMAJ{ (ng/mLl AVCo-oo (ng.h/mLl tll2 (hl MRT (hl
Tyrosol (after administration of salidroside) 1. 794 1.000 1081.154 1185.491 2.074 2.993
Tyrosol (after administration of SHR-5) 0.958 2.000 159.3400 334.117 2,005 2.982
DISCUSSION The pharmacokinetic parameters of rhodioloside determined following administration of the pure compound and as a component of the special extract (SHR-5) of R. rosea, are compared in Table 4. Most ofthe measured parameters differed significantly according to the manner in which rhodioloside was administered: thus the maximal concentration of rhodioloside in blood plasma was attained later (tMAX ) when the pure compound was given, and the values of CMAX ' AUC o_v' tl/2 and MRT were
326
Compo Bio. Nut. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
some 1.2-2.0 times lower than those determined when the compound was administrated as SHR-5. In contrast, the rate of distribution ofrhodioloside within tissues was almost the same since the values of VDSS determined following administration of pure rhodioloside and SHR-5 were similar. The differences outlined above could be accounted for by the presence in SHR-5 of compounds which are chemically similar to rhodioloside and which compete with rhodioloside for the same metabolic enzymes. If this suggestion is correct, it implies that the content oftyrosol in blood plasma should be higher after administration of pure rhodioloside than after administration of SHR-5. Comparison of the pharmacokinetics of tyrosol following administration of pure rhodioloside and of SHR-5 (Fig 7) shows that the initial concentration of tyrosol in blood plasma was significantly higher (particularly in the first 2 h) after administration of the pure compound, but then decreased at approximately the same rate for both forms of administration. At the same time the values for CMAX and AUC o_oo of tyro sol are significantly higher after administration ofrhodioloside, but the values of the elimination parameters, such as tl/2 and MRT, are almost the same (Table 6). It may thus be concluded that the biotransformation of rhodioloside was more rapid following oral administration of the pure compound than after administration of SHR-5. Moreover, the maximum concentration of tyrosol was detected after only 1 h following intravenous administration ofSHR-5 indicating that tyro sol detected in blood originated not only from the free tyrosol in SHR-5 but also from other precursors present in SHR-5. The above results indicate that the biotransformation ofrhodioloside occurs within the first 2 h following administration of the compound either in pure form or as SHR-5 but, when administered as a component of the extract, its metabolism takes place more slowly because ofthe presence of other compounds such as rosavine, rosarine, rosine etc, which compete for the metabolising enzymes and hence decrease the rate of formation of tyrosol from rhodioloside. It can be seen from Fig 7 that the maximum concentration of tyro sol in blood plasma was observed 1 h after multiple administration ofSHR-5 at doses of 20 and 50 mg/kg: after 1-1.5 h the concentration fell sharply and then decreased exponentially. Whilst within the first 2 h following the fifth administration ofSHR-5 the concentration oftyrosol in blood was significantly higher than the concentration ofrhodioloside, the levels of the two analytes in blood plasma became almost the same at longer time periods.
These results indicate that the rate of biotransformation of rhodioloside is practically the same following single or multiple regimes of administration ofthe drug, and that component ofSHR-5 cannot induce the activation of metabo Ii sing enzymes. These results also help to explain why
Pharmacokinetics of Active Constituents of Rhodiola rosea
327
there are no statistically significant differences in the phannacological effects ofrhodioloside and tyrosol, such as in the increase of proteolytic activity of muscles or in alterations oflevels of ATP, ADP, AMP etc, after exhausted physical loading (Saratikov, 1973). The low bioavailability of rosavin (20 - 26%) means that the native compound cannot be detected in blood plasma after multiple administration of SHR-5 at a dose of 20 mg/kg. This suggests that rosavin is rapidly metabolised, perhaps during the first passage through the liver. The low bioavailability of rosavin may also explain why some effects of this compound are observed only when it is administrated in very high doses (50 mg/kg in mice) and not with lower doses (3 and 10 mglkg in mice) corresponding to single therapeutic and daily human doses of SHR-5 (Sokolov et al., 1990). The pharmacokinetics ofrosavin in humans is quite different to that in rats. For example, both t max and elimination rate are longer in humans. A similar tendency is also seen in the pharmacokinetics of rhodioloside, where it is better available in the blood of patients than rosavin. For example, the ratio of AUC o_oo Idose is nearly twice as high for rhodioloside, in comparison to rosavin (Table 5). As a matter of fact, the ratio of elimination halftimes has the exact same value of 1.8 (Table 5). This clearly indicates that there is a higher, and thus longer-lasting, concentration of rhodiolside in the blood, in comparison to rosavin. These data favours rhodioloside as the major biologically active constituent of Rhodiola SHR5 extract. It is noteworthy to mention the beneficial effect of Rhodiola rosea on mental performance in humans, which was observed one h after oral administration and lasted for more than three h. During this time period, it was observed that the concentration ofrhodioloside in human blood was about 587 ± 102 ng/mL (after 1 h) and 483 ± 102 ng/mL (after 4 h) (Fig 8).
ACKNOWLEDGEMENTS This study was sponsored by Swedish Herbal Institute, Gothenburg, Sweden.
REFERENCES Barnaulov, O.D., Limarenko, A.Y., Kurkin, V.A., Zapesochnaya, G.G. and Shavlinskii, A.N. (1986). Comparative evalution of biological activity of compounds, isolated from Rhodiola L. speicies. Kh~m. Farm. Zh., 20(9): 1107-1112. [Article in Russian] Brichenko, V.S., Kupriyanova, I.E. and Skorokhodova, T.F. (1986). The use of herbal adaptogens together with tricyclic anti-depressants in patients with psychogenic depressions. In: Modern problems of pharmacology and search for new medicines, Vol 2, Ed. By Goldsberg, E.D., Tomsk State University Press, Russia, pp. 58-60. Darbinyan, V., Kteyan, A., Panossian, A., Gabrielian, E., Wikman, G. and Wagner, H. (2000). Rhodiola rosea in stress-induced fatigue: a double-blind cross-over study of a standardised extract SHR-5 with a repeated low-dose regimen on the mental performance of healthy physicians during night duty. Phytomedicine, 7: 365-371.
328
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Darbinyan, V., Aslanyan, G., Amroyan, E., Gabrielyan, E., Malmstrom, C. and Panossian, A. (2007). Clinical trial of Rhodiola rose a L. extract SHR-5 in the treatment of mild to moderate depression. Nord J Psychiatry, 61: 343-8. Krasik, E.D., Petrova, KP. and Rogulina, G.A. (1970). About the adaptogenic and stimulating effect of Rhodiola rosea extract. In: Proceedings of all-union and 5th Sverdlovsk area conference of neurobiologists, psychiatrists and neurosurgeons, Ed. By Avrutskiy, G.Y., Sverdlovsk Press, Russia, pp. 215-217. Linh, P.T., Kim, Y.H., Hong, S.P., Jian, J.J. and Kang, J.S. (2000). Quantitative determination of salidroside and tyrosol from the underground part of Rhodiola rosea by high performance liquid chromatography. Arch. Pharm. Res. 23: 349-52. Mikhailova, M.N. (1983). Clinical and experimental substantiation of asthenic conditions therapy using Rhodiola rosea extract. In: Current problems of psychiatry, Ed. By Goldsberg, E.D., Tomsk State University Press, Russia, pp. 126-127. Miro-Casas, M.E., Farre Albaladejo, M.F., Covas, M.l., Colomer, M., Lamuela Raventos, R.M. and de la Torre Fornell, R. (2001a). Tyrosol bioavailability in humans after ingestion of virgin olive oil. Clin. Chem, 47: 341-343. Miro-Casas, M.E., Farre Albaladejo, M.F., Covas M.l., Rodriguez, J.O., Colomer, E.M., Lamuela Raventos, R.M. and de la Torre, R. (2001b). Capillary gas chromatographymass spectrometry quantitative determination of hydroxytyrosol and tyrosol in human urine after olive oil intake. Anal Biochem, 294(1): 63-72. National pharmacopoeia of the USSR (1990). National pharmacopoeia of USSR XI, Vol 2. Meditsina, Moscow, 364 pp Olsson, E.M.G., von Scheele, B. and Panossian, A.G. (2009). A randomized double-blind placebo controlled parallell group study of SHR-5 extract of Rhodiola rosea roots as treatment for patients with stress related fatigue. Planta medica, 75:105-12. Panossian, A., Hovhannisyan, A., Abrahamyan, H., Gabrielyan, E. and Wikman, G. (2002). Pharmacokinetics of rhodioloside and rosavin from standardized Rhodiola rosea extract SHR-5 in rats. Pharmacologist, 44(2): AI71, Supp.1. Panossian, A., Hambartsumyan, M., Hovanissian, A, Gabrielyan, E. and Wikman, G. (2007). The Adaptogens Rhodiola and Schizandra modify the response to immobilization stress in rabbits by suppressing the increase of phosphorylated stress-activated protein kinase, nitric Oxide and cortisol. Drug Targets Insights, 1: 39-54. Panos sian, A., Nikoyan, N., Ohanyan, N., Hovhannisyan, A., Abrahamyan, H., Gabrielyan, E. and Wikman, G. (2008). Comparative study of Rhodiola preparations on behavioral despair of rats, Phytomedicine, 15(1): 84-91. Ritschel, W.A. and Kearm, G.L. (998). Handbook of basic pharmacokinetics. 5th Edn., Drug Intelligence Publication, Hamilton. Saratikov, A.S., Marina, T.F. and Kaliko, l.M. (1965). The stimulating effect of Rhodwla rosea on the higher brain structures. Newsletter Sib Branch USSR Acad Sci., 8: 120-125 Saratikov, A.S., Krasnov E.E., Chnikina L.A., Duvidson L.M., Sotova M.I., Marina T.F., Nechoda M.F., Aksenova R.A. and Tschrdinzeff S.G. (1968). Rhodioloside in neues Glykosid aus Rhodwla rosea und seine pharmakologischen Eigenschaften. Die Pharmazie, 23: 392-395. Saratikov, A.S. (1973). The golden root
Pharmacokinetics of Active Constituents of Rhodiola rosea
329
Spasov, A.A., Mandrikov, V.B. and Mironova, LA. (2000a). The effect of the preparation rhodiosin on the psychophysiological and physical adaptation of students to an academic load. Eksp Klin Farmakol, 63: 76-78. Spasov, A.A., Wikman, G.K., Mandrikov, V.B., Mironova, LA. and Neumoin, V.V. (2000b). A double-blind, placebo-controlled pilot study of the stimulating and adaptogenic effect of Rhodiola rosea SHR-5 extract on the fatigue of students caused by stress during an examination period wiyh a repeated low-dose regimen. Phytomedicine, 7(2): 85-89. Visioli, F., Galli, C., Bornet, F., Mattei, A., Patelli, R., Galli, G. and Caruso, D. (2000). Olive oil phenolics are dose-dependently absorbed in humans. FEES Letters, 468: 159-160.
"This page is Intentionally Left Blank"
17 Metabolism and Pharmacokinetic Studies of Coumarins BEHERA D. 1 AND AnDEPALLI V.l,*
ABSTRACT Coumarin is a naturally occurring compound found in a wide variety of plants, microorganisms and in some animal species, with multiple uses. The metabolism, toxicity and results of tests for carcinogenicity of coumarins present in foodstuffs and cosmetic products have been reviewed with respect to safety for humans. Coumarin is a natural product, which exhibits marked species differences in both metabolism and toxicity. Coumarin is metabolized mainly by two important pathways: 7-hydroxylation and metabolism of the lactone ring which involves ring opening and the cleavage carbon atom 2 to yield CO 2 (also known as 3,4-epoxidation). While 7-hydroxylation is the major pathway of coumarin in most subjects, humans can also metabolize coumarin by the 3,4-epoxidation and possibly other pathways to various other metabolites. In contrast, the major route of coumarin metabolism in the rat and mouse is by 3,4-epoxidation pathway resulting in the formation of toxic metabolites. These Phase I metabolites are further conjugated either with glucuronate, sulfate, acetate and amino acids (Phase II reactions). Coumarin metabolism is catalyzed by various cytochrome P450 (CYP) isoforms in humans and other species, thus showing marked species differences. There is an extensive literature available on the disposition and metabolism of coumarin in a range of animal species and humans. Both in vitro and in vivo metabolic studies have been performed. Studies have shown that coumarins are metabolized in the body by hepatic as well as extrahepatic organs. In one of our in vitro research study, two coumarin 1. Dept. of Pharmacology, School of Pharmacy & Technology Management, NMIMS
*
University, Vile-Parle(W), Mumbai - 400 056, Maharashtra, India. Corresponding author: E-mail: [email protected]
332
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
derivatives, 7-ethoxy coumarin (7-EC) and 7-hydroxy coumarin (7-HC) showed significant degradation in rat and mice hepatic as well as extrahepatic (intestine, kidneys, lungs and brain) subcellular fractions, thus showing the relative contribution of hepatic and extra-hepatic metabolism to total clearance of both drugs, being remarkably high, ranging from 62% to 77% and 22% to 38% respectively of the total metabolic clearance in rats. The majority of tests for mutagenic and genotoxic potential suggest that coumarin is not a genotoxic agent. The target organs for toxicity and carcinogenicity in the rat and mouse are primarily the liver and lung. Moreover, the dose-response relationships for coumarin-induced toxicity and carcinogenicity are non-linear, with tumour formation only being observed at high doses which are associated with hepatic and pulmonary toxicity. The maximum daily human exposure to coumarin from dietary sources for a 60 kg consumer has been estimated to be 0.02 mg/kg/day. From fragrance use in cosmetic products, coumarin exposure has been estimated to be 0.04 mg/kg/day. The total daily human exposure from dietary sources together with fragrance use in cosmetic products is thus 0.06 mg/kg/day. No adverse effects of coumarin have been reported in susceptible species in response to doses, which are more than 100 times the maximum human daily intake. The mechanism of coumarin-induced tumour formation in rodents is associated with metabolism-mediated, toxicity and it is concluded that exposure to coumarin from food and/or cosmetic products poses no health risk to humans. Key words: Coumarin, metabolism, pharmacokinetics, genotoxicity, carcinogenicity, species differences
INTRODUCTION Coumarin (l,2-benzopyrone; coumarinic anhydride) is a white crystalline solid (molecular weight 146.15, melting point 68-70°C, boiling point 297299°C), freely soluble in ethanol, chloroform, diethyl ether and oils, but slightly soluble in water (Cohen, 1979). The majority of coumarin used commercially is synthesized from salicylaldehyde, although high-grade coumarin is still isolated from tonka beans (DeGarmo & Raizman, 1967; Hawley, 1971). As described below, coumarin exhibits marked species differences in both metabolism and toxicity. The objective of this review which incorporates our own research findings on Drug Metabolism and Pharmacokinetics (DMPK) aspects of two coumarin derivatives is to evaluate data on coumarin metabolism. Further, a general discussion has been made on toxicities with respect to the hazards caused by this compound in humans, arising from its presence in food and fragrance products.
Metabolism and Pharmacokinetic Studies of Coumarins
333
Uses of Coumarin Coumarin is used as a fixative and enhancing agent in perfumes and it is also used as one of the ingredients in toilet soap, detergents, toothpaste, tobacco products and some alcoholic beverages (Cohen, 1979; Opdyke, 1974). Large quantities of coumarin are also used in rubber and plastic materials and in paints and sprays to mask unpleasant odours (Fentem & Fry, 1993). Coumarin has also been used in clinical medicine because of its ability to activate macrophages that underlies its use for the treatment of high protein oedema and its immunomodulatory properties used in the treatment of brucellosis (Egan et at., 1990). The use of coumarin as a food flavour was banned in USA in 1954 based on reports of hepatotoxicity in rats, prior to the existence of any carcinogenicity and mutagenicity data and was recommended for withdrawal from use in the UK in 1965 (Cohen, 1979; Opdyke, 1974).
Occurrence and Human Exposure Coumarin is a naturally occurring compound, found in a wide variety of plants, microorganisms and in some animal species (Feuer, 1974; Soine, 1964). It was first isolated from tonka beans, and is found at high levels in some essential oils, particularly cinnamon bark oil (7000 ppm), cassia leaf oil (up to 87,300 ppm), peppermint oil (20 ppm) and lavender oiL Coumarin is also found in fruits (e.g. bilberry, cloudberry), green tea and other foods, such as chicory (TNO, 1996). Many coumarin derivatives have also been found in plants, being present in free state and as glucosides. Humans are exposed to coumarin from food, beverages, caramel confectionery, chewing gum and alcoholic beverages. They are also exposed to coumarin from fragrance use in cosmetic products (Cohen, 1979; Opdyke, 1974). Such products include antiperspirant deodorants, bath products, body lotions, face creams, fragrance creams, hair sprays, shampoos, shower gels and toilet soaps. Overall, a realistic total daily human exposure to coumarin from the diet and from fragrance use in cosmetic products would be 0.06 mg/kg/day.
Pharmacokinetics
of Coumarin
Extensive literature exists on the disposition and metabolism of coumarin in a range of animal species and humans. Both in vitro and in vivo metabolic studies have been performed in animal species.
Absorption, Distribution and Excretion Following oral administration, it was found that coumarin was rapidly absorbed from the gastrointestinal tract and distributed throughout the body (Cohen, 1979; Fentem & Fry, 1993; Pelkonen et at., 1997). The compound appears to be extensively metabolized in all species with little
334
Comp. Bio. Nat. Prod., VoL 2 - Efficacy, Safety & Clinical Evaluation I
unchanged coumarin being excreted. No evidence of significant tissue accumulation of coumarin and/or coumarin metabolites was obtained after oral administration to rats and rabbits or intraperitoneal (ip) administration to rats CKaighen & Williams, 1961; van Sumere & Teuchy, 1971). Important quantitative differences exist between species in the routes of elimination of coumarin metabolites. The majority of studies have demonstrated a relatively large amount of biliary excretion in the rat with an appreciable proportion of the dose being excreted in the faeces. After a 50 mg/kg oral or ip dose of coumarin to rats some 50% of the dose was excreted in the bile as unknown metabolites within 24 h (Williams et al., 1965). The urine appears to be the major route of coumarin metabolite excretion in species such as the Syrian hamster, rabbit and baboon. The rapid excretion of coumarin, primarily as 7-hydroxycoumarin (7-HC), in the urine of human subjects given coumarin suggests that there is little or no biliary excretion of coumarin metabolites in humans (Shilling et al., 1969). The pharmacokinetics of coumarin has been studied in a number of species including the rat (Hardt & Ritschel, 1983; Piller, 1977; Ritschel & Hussain, 1988), dog (Ritschel & Grummich, 1981), gerbil (Ritschel & Hardt, 1983), rhesus monkey (Ritschel et al., 1988) and in man. The elimination half-life (t 1J2 ) of coumarin was found to be similar in all species examined, being around 1-2 h in humans and between 1 and 4 h in other species. In the rat, following an ip dose between 2.5 and 60 mg/kg, coumarin was rapidly eliminated from whole blood with an elimination half-life varying from 1 to 3 h (Hardt & Ritschel, 1983). In a later study conducted in rat, it was found that coumarin was rapidly eliminated from the systemic circulation (t 1J2 less than 1 h) after an intravenous (iv) dose of 1 mg/kg and 20% of an oral dose of coumarin (1 mg/kg) was absorbed intact due to extensive first-pass metabolism (Ritschel & Hussain, 1988). In the gerbil, coumarin at an ip dose of 40 mg/kg, was rapidly absorbed into the blood, and eliminated with a half-life of approximately 1 h (Ritschel & Hardt, 1983), whereas in the dog, the elimination of coumarin following an iv dose of 0.25 or 0.5 mg/kgwas slower, the t1J2 being 2.7 and 3.7 h, respectively (Ritschel & Grummich, 1981). In contrast to the rat, approximately 45% of an oral dose of coumarin of 1 mg/kg was absorbed intact in the dog. The pharmacokinetics of coumarin in the rhesus monkey were found to be similar to that in the dog (Ritschel et al., 1988), with oral bioavailability approximately 45% after an oral dose of 1 mg/kg and elimination half-life approximately 2 h. Pharmacokinetic studies in humans have demonstrated that coumarin is completely absorbed from the gastrointestinal tract after oral administration and extensively metabolized by the liver in the first-pass, with only between 2 and 6% reaching the systemic circulation intact
Metabolism and Pharmacokinetic Studies of Coumarins
335
(Ritschel et al., 1977, 1979; Ritschel & Hoffman, 1981). The elimination of coumarin from the systemic circulation is rapid, the half-lives following iv doses of 0.125, 0.2 and 0.25 mg/kg being 1.82, 1.46 and 1.49 h, respectively (Ritschel et al., 1976).
Coumarin Metabolism Pathways The metabolism of coumarin has been studied in vivo and in vitro in a wide range of species including humans. In vitro models employed include tissue slices, hepatocytes, subcellular fractions, and purified and c-DNAexpressed enzymes. Coumarin may be metabolized by hydroxylation at all six possible positions (i.e. carbon atoms 3, 4, 5, 6, 7 and 8) to yield 3-, 4-, 5-, 6-, 7- and 8-hydroxycoumarins (3-, 4-, 5-, 6-, 7- and 8-HCs) and by opening of the lactone ring to yield various products including 0hydroxyphenylacetaldehyde (o-HPA), o-hydroxyphenylethanol (o-HPE), 0hydroxyphenylacetic acid (o-HPAA) and o-hydroxyphenyllactic acid (0HPLA). Additional metabolites of coumarin include 6,7 -dihydroxycoumarin (6,7-diHC), o-coumaric acid (o-CA), o-hydroxyphenylpropionic acid (o-HPPA) and dihydrocoumarin (DHC). The known pathways of coumarin metabolism are summarized in Fig 1.
I
o-CA +------. o-HPPA
3-HC---.o-HPLA
1
r
4-HC
~
I
Coumarin---+o-HPE -----+ o-HPAA
5-,6- and a-H?,.------:
J
\
1
7-EC
6,7-';HC
~HPE
7-HC
/~
7-HC-glucuronide
7-HC-sulfate
Fig 1. Some pathways of coumarin metabolism (Born et al., 1997c; Cohen, 1979; Fentem et al., 1991a; Lake et at., 1992a, b; Norman & Wood, 1984). Abbreviations used are o-coumaric acid (o-CA), dihydrocoumarin (DHC), 6,7-dihydroxycoumarin (6,7-diHC), hydroxycoumarin (HC), o-hydroxyphenylacetaldehyde (o-HPA), 0hydroxyphenylethanol (o-HPE), o-hydroxyphenylaceticacid (o-HPAA), 0hydroxyphenyllactic acid (o-HPLA), o-hydroxyphenylpropionic acid (o-HPPA)
The two important pathways of coumarin metabolism are 7hydroxylation and metabolism of the lactone ring which involves ring
336
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
opening and the cleavage of carbon atom 2 to yield carbon dioxide. The latter pathway is often referred to as the "3-hydroxylation" pathway of coumarin metabolism (Cohen, 1979; Fentem & Fry, 1993; Kaighen & Williams, 1961; Lake et al., 1989a; Pelkonen et al., 1997).
In vivo Metabolism Studies Early studies into the in vivo metabolism of coumarin were conducted by Booth et al. (1959) and by Mead et al. (1958a, b) using unlabelled materiaL Booth et al. (1959) reported that coumarin was metabolized to o-HPAA in both the rat and rabbit, whereas only the rabbit appeared to excrete appreciable quantities of 3-HC and 7-HC. Mead et al. (1958a) observed that coumarin was converted to 3-HC, 7-HC and 8-HC in the rat and rabbit, with 3-HC, 5-HC, 7-HC and 8-HC being formed in the mouse, guinea pig and ferret. Additional studies with 3-HC, 4-HC, 5-HC, 6-HC, 7-HC and 8HC in the rabbit demonstrated that all six hydroxycoumarins were excreted in the urine as glucuronide conjugates and that all except 4-HC were also excreted as sulfate conjugates (Mead et al., 1958b). Generally, studies with 7-HC in several other species (Cohen, 1979) have demonstrated that if formed, this coumarin metabolite is extensively conjugated with either Dglucuronic acid and/or sulfate. Coumarin was also extensively metabolized in the rat with only small amounts of unchanged coumarin being detected in the excreta. Various metabolites including o-HPAA were detected in the faeces, with urinary 3HC, 7-HC, o-HPLA and o-HPAA accounting for 1.8, 0.4, 0.8 and 20% of the dose, respectively (Kaighen & Williams, 1961). The metabolism of [2_14CJ coumarin in the rat administered by the ip route was studied by van Sumere and Teuchy (1971). In this study a large percentage of the label was excreted in the expired air (i.e. Oas 14C0 2), because carbon atom 2 is removed when the lactone ring is opened to form 0- HPA and subsequent metabolites (see Fig 2). Analysis of the urine revealed a number of coumarin metabolites including 5-HC, 7-HC, 8-HC, o-CA, o-HPAA and o-HPAA (van Sumere & Teuchy, 1971). Clear species differences in the extent of coumarin metabolism to o-HPAA have been reported. For example, o-HPAA appears to be a major urinary metabolite in the marmoset, but only a minor metabolite in the baboon (Lake et al., 1989b; Waller & Chasseaud, 1981). Apart from glucuronide and sulfate conjugates of hydroxycoumarins, some other phase II coumarin metabolites have been identified. For example, 0CA may be conjugated with glycine (Feuer, 1974). There is some evidence that coumarin may be metabolized by the gastrointestinal microflora (Cohen, 1979). For example, Scheline (1968) reported that rat caecal microflora could convert coumarin to DHC and 0HPPA.
Metabolism and Pharmacokinetic Studies of Coumarins
337
Because of the relative ease of measurement of 7-HC, many in vivo studies have examined this pathway of coumarin metabolism and marked species differences have been reported. Overall, several species including the rat, most mouse strains, Syrian hamster, guinea pig, ferret, dog, marmoset and squirrel monkey appear to be poor 7-hydroxylators of coumarin excreting 5% or less of the administered dose as urinary 7-HC. Certain mouse strains, such as DBAl2 and 129IRr strains, which have relatively high hepatic coumarin 7-hydroxylase activity, excrete up to 26% of an ip administered dose of coumarin as 7-HC (Lush & Andrews, 1978). Species such as the rabbit, cat and pig have been reported to excrete 1219% ofthe dose as urinary 7-HC (Gangolli et al., 1974; Kaighen & Williams, 1961), whereas in two studies the baboon, an Old World primate, has been reported to be an extensive 7-hydroxylator of coumarin with 60-66% of the dose being rapidly excreted in the urine as 7-HC (Gangolli et al., 1974; Waller & Chasseaud, 1981). In the majority of human subjects studied coumarin is extensively metabolised to 7-HC. The measurement of urinary 7-HC following an oral dose of coumarin has been employed as a biomarker of human hepatic CYP2A6, the cytochrome P-450 (CYP) isoform which is responsible for coumarin 7-hydroxylation in human liver. While 7-hydroxylation is the major metabolic pathway of coumarin in most subjects, humans can also metabolise coumarin by the 3,4-epoxidation and possibly other pathways to o-HPAA. In a dermal administration study conducted in three subjects the 0-12 h urine contained 51% of the dose (range 44-57%) as total (free and conjugated) 7-HC, together with 1% (range 0-1.5%) of the dose as 0HPAA and 2.8% (range 2.1-3.5%) of the dose as other unknown metabolites (Huntingdon Life Sciences, 1996a). The marked inter-individual variation in coumarin metabolism to 7HC has led to studies to evaluate whether a genetic polymorphism exists in human CYP2A6. Work by Fernandez-Salguero et al. (1995) demonstrated variant alleles in the human CYP2A6 gene. Three alleles were identified namely the wild type, CYP2A6v1 and CYP2A6v2, which are now designated CYP2A6*1, CYP2A6*2 and CYP2A6*3, respectively (Gullsten et al., 1997; Hadidi et al., 1997). The relationship between CYP2A6 gene polymorphism and risk of liver cancer and cirrhosis has been recently investigated (Gullsten et al., 1997). CYP2A6 function in vivo, as assessed by the urinary 7-HC excretion test, is impaired in patients with alcoholic liver disease (Sotaniemi et al., 1995) and in both adults and children by hepatitis A infection (Pasanen et al., 1997). In contrast, compared with normal subjects, urinary 7-HC excretion is enchanced in epileptic patients (Sotaniemi et al., 1995). Such data suggest that anticonvulsant drugs such as carbamazepine and phenobarbitone can induce levels of hepatic CYP2A6 £n vwo.
338
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
In vitro Metabolism Studies Early studies demonstrated that coumarin was metabolized in rat liver microsomes by CYF-dependent pathways and that a number of metabolites were formed including 3-HC, o-HPLA and o-HPAA (Feuer 1970; Gibbs et at., 1971). Depending on the coumarin substrate concentration employed, o-HPA is a major metabolite of coumarin in liver microsomes from several species (Fentem & Fry, 1992; Fentem et at., 1991a; Lake et ai., 1992a, b; Peters et at., 1991). Other coumarin metabolites that may be observed in incubations with liver microsomes include 4-HC, 5-HC, 6-HC, 7-HC, 8HC, 6,7-diHC, o-CA and o-HPPA. Depending on the coumarin substrate concentration employed, o-HPA is a major metabolite of coumarin in liver microsomes from several species (Fentem & Fry, 1992; Fentem et at., 1991a; Lake et at., 1992a, b; Peters et at., 1991). Other coumarin metabolites that may be observed in incubations with liver microsomes include 4-HC, 5HC, 6-HC, 7-HC, 8-HC, 6,7-diHC, o-CA and o-HPPA. Many studies have examined coumarin 7 -hydroxylase activity, determined by fluorimetric or other procedures, in hepatic microsomal preparations (Cohen, 1979; Fentem & Fry, 1993; Pelkonen et at., 1993, 1997). Rat liver microsomes have little or no coumarin 7-hydroxylase activity (Bogan et ai., 1996; Bullock et at., 1998; Creaven et at., 1965; Dominguez et at., 1990; Fentem & Fry, 1991, 1992; Lake et at., 1989b, 1992a; Pearce et at., 1992; Pelkonen et at., 1985, 1993; Raunio et at., 1988b; Walters et at., 1980). In contrast, coumarin 7-hydroxylase activity is readily detectable in liver microsomes from other species including the mouse, gerbil, guinea pig, rabbit and human (Cohen, 1979; Fentem & Fry, 1993; Pelkonen et at., 1993, 1997). In keeping with in vivo studies, marked strain differences have been reported in mouse hepatic microsomal coumarin 7 -hydroxylase activity. Negishi et at. (1989) observed higher enzyme activity in female than in male 129/J strain mice. Other studies have also demonstrated both sex and strain differences in mouse coumarin 7-hydroxylase, with enzyme activity in high activity strains, such as DBAl2 and 129, being greater in female than in male mice (Lovell et at., 1999; van Iersel et at., 1994a). The CYF isoform responsible for coumarin 7-hydroxylation in mouse liver is CYF2A5 (Chang & Waxman, 1996; Honkakoski & Negishi, 1997; Pearce et at., 1992; Pelkonen et at., 1993, 1997). In line with the sex difference in coumarin 7-hydroxylase activity, the expression of CYF2A5 is female predominant in mouse liver (Chang & Waxman, 1996; Honkakoski & Negishi, 1997; Pearce et at., 1992; Squires & Negishi, 1990). Like mouse CYF2A5 and human CYF2A6, but unlike rat CYF2A1 and CYF2A2 and mouse CYF2A4, Syrian hamster CYF2A8, rabbit CYF2A10 and CYF2A isoforms from the baboon and cynomolgus monkey are all able to efficiently
Metabolism and Pharmacokinetic Studies of Coumarins
339
catalyse the 7-hydroxylation of coumarin (Chang & Waxman, 1996; Honkakoski & Negishi, 1997). While much attention has focused on the measurement of coumarin 7-hydroxylase activity in liver microsomes, it should be noted that this is only a minor pathway of coumarin metabolism in many species. Coumarin 7-hydroxylation also only represents a fraction oftotal coumarin metabolism in gerbil liver microsomes (Fentem & Fry, 1992). Moreover, in agreement with in vivo data (Lush & Andrews, 1978), coumarin 7-hydroxylation is not the major pathway of coumarin metabolism even in mouse strains with relatively high coumarin 7-hydroxylase activity (Lovell et al., 1999). Many studies have investigated coumarin 7-hydroxylase activity in human liver microsomes (Chang & Waxman, 1996; Forrester et al., 1992; Kratz, 1976; Maurice et al., 1991; Miles et al., 1990; Pearce et al., 1992; Pelkonen et al., 1993, 1997; Shimada et al., 1996; van Iersel et al., 1994b; Wrighton & Stevens, 1992; Yamano et al., 1990; Yun et al., 1991). The major cytochrome P-450 isoform responsible for coumarin 7-hydroxylation in human liver is CYP2A6 (Chang & Waxman, 1996; Honkakoski & Negishi, 1997; Pelkonen et al., 1993, 1997), whereas another CYP2A sub-family isoform present in human liver (Nelson et al., 1996), namely CYP2A7, does not possess coumarin 7-hydroxylase activity (Ding et al., 1995; Yamano et al., 1990). Compared to some other CYP isoforms, such as CYP1A2, CYP2C and CYP3A isoforms, levels of CYP2A6 are relatively low in human liver (Shimada et al., 1994, 1996; Wrighton & Stevens, 1992). In one study with liver micro somes from 30 Caucasian and 30 Japanese subjects, CYP2A6 was reported to be 4.0% of the total cytochrome P-450 content (Shimada et al., 1994). Unlike mouse liver CYP2A5, there appears to be a lack of any sex difference in levels of CYP2A6 in human liver (Chang & Waxman, 1996). Other studies suggest that CYP2A6 may be induced by various agents and also inhibited by known inhibitors of CYP isoforms (Chang & Waxman, 1996). For example, levels of CYP2A6 in cultured human hepatocytes are increased by treatment with phenobarbitone, dexamethasone and rifampicin, but not by b-naphthoflavone and pyrazole (Chang & Waxman, 1996; Dalet-Beluche et al., 1992; Maurice et al., 1991). Many studies have demonstrated marked interindividual variation in levels ofCYP2A6 protein, mRNA and associated coumarin 7-hydroxylase activity in human liver microsomes (Chang & Waxman, 1996; Forrester et al., 1992; Maurice et al., 1991; Miles et al., 1990; Pearce et al., 1992; Pelkonen et al., 1993, 1997; Shimada et al., 1996; van Iersel et al., 1994b; Wrighton & Stevens, 1992; Yamano et al., 1990; Yun et al., 1991). While most studies with human liver microsomes have focused on 7-hydroxylation, the formation of other coumarin metabolites, including products of the 3,4epoxidation and other pathway(s) (i.e. 3-HC, o-HPA, o-HPE and o-HPAA),
340
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
4-HC, 5-HC, 6-HC, 8-HC, 6,7-diHC, o-CA and o-HPPA have been observed (Fentem & Fry, 1992; Lake et al., 1992a; van Iersel et al., 1994b). Apart from subcellular fractions the in vitro metabolism of coumarin has also been studied in hepatocytes and precision-cut liver slices from various species, and in precision-cut rat lung slices (den Besten et al., 1990; Donato et al., 1998; Lake et al., 1995; Price et al., 1995; Ratanasavanh et al., 1996; Steensma et al., 1994). Generally, the species differences in coumarin metabolism observed in hepatocytes and liver slices are in agreement with available literature data from in vivo studies and from investigations with liver microsomes. In our research study, two coumarin derivatives, 7-ethoxy coumarin (7-EC) and 7-hydroxy coumarin (7-HC) were chosen as model compounds to study hepatic and extra-hepatic (intestine, kidneys, lungs and brain) metabolism in mice and rat tissue subcellular (S9 and microsomal) fractions and to scale the observed in vitro clearance to in vivo plasma clearance in rats. It was found that, both drugs showed significant metabolic degradation in rat liver subcellular fractions as compared to subcellular fractions obtained from intestine, kidney, lung and brain. The total in vitro metabolic clearance for 7-EC and 7-HC was determined by adding the individual in vitro organ clearance values obtained in hepatic and extra-hepatic microsomes or S9 fractions. The predicted in vivo clearance for 7-HC was 63.10 and 81.35 mUmin/kg by in vitro scaling from micro somes and S9 fractions, respectively. For 7-EC, the values were 78.36 and 76.72 mU min/kg, respectively. The predicted clearance was found to be reasonably accurate with slight over and underprediction. Interestingly, the relative contribution of hepatic and extra-hepatic metabolism to total clearance of 7-EC and 7-HC was remarkably high, ranging from 62% to 77% and 22% to 38%, respectively, of the total metabolic clearance. It is concluded that, the model of multi-organ subcellular fractions is a useful in vitro tool for prediction of in vivo metabolic clearance, as it can provide information about the relative contribution of extra-hepatic metabolism in addition to hepatic metabolism to the total metabolic clearance (Behera et al., 2008).
Genotoxicity Studies A number of studies have examined the mutagenic and genotoxic potential of coumarin. Overall, the data suggest that coumarin is not a genotoxic agent. No evidence for coumarin-induced genotoxicity has been observed in in vivo studies (Drosophila melanogaster and mouse micronucleus tests). The findings from these in vivo studies are supported by data from some, but not all, of the in vitro studies. In in vivo studies coumarin did not induce sex-linked recessive lethal mutations in germ cells of male D. melanogaster exposed as adults either by feeding or by injection (NTP, 1993a; Yoon et al., 1985), or as larvae by feeding (NTP, 1993a; Valencia et al., 1989). The administration of65 and 130 mg/kg/day coumarin by gavage
Metabolism and Pharmacokinetic Studies of Coumarins
341
for 7 days did not induce micronuclei in the bone marrow of male and female immunocompromised (ICR) mice (Morris & Ward, 1992). A number of in vitro genotoxicity studies have been conducted with coumarin. Recently the effect of coumarin on unscheduled DNA synthesis (UDS) in cultured precision-cut human liver slices has been evaluated. Previous work has demonstrated that like hepatocytes, cultured liver slices may also be employed to screen xenobiotics for effects on UDS (Lake et al., 1996). No evidence for coumarin-induced UDS was obtained in experiments with cultured liver slices from four subjects employing coumarin concentrations of up to 5 mm (Beamand et al., 1998). Some cytotoxicity was observed at high coumarin concentrations and the functional viability of the human liver slice preparations was confirmed by parallel studies with known genotoxins. Coumarin has also been reported not to induce UDS in a rat tracheal epithelium culture system (Ide et al., 1981). No evidence of mutagenicity was obtained when coumarin was examined in S. typhimurium strains TA98, TA1535, TA1537 and TA1538, both with or without metabolic activation according to Rhodia studies. The ability of coumarin to induce sister chromatid exchanges and chromosomal aberrations in Chinese hamster ovary (CHO) cells has been investigated for the NTP (Galloway et al., 1987). Like certain other natural products, coumarin can have antimutagenic properties (Brown et al., 1981; Edenharder & Tang, 1997; Grigg, 1977; Imanishi et al., 1990; Sanyal et al., 1997; Sasaki et al., 1987, 1990). Coumarin has been reported to induce micronuclei in HepG2 cells at high (500 mg/mL) concentrations, but to protect against the effects of known chemical mutagens (namely 2-amino3-methylimidazo[4,5-flquinoline (IQ) and other cooked food mutagens) at low (.::;5 f.lg!mL) concentrations (Sanyal et al., 1997). Coumarin has also been shown to suppress UV- and X-ray-induced 6-thioguanine resistant mutations in Chinese hamster V79 cells CImanishi et al., 1990). Brown et al. (1981) reported that coumarin decreased the mutagenicity of benzo[a]pyrene in the Ames test, whereas Edenharder and Tang (1997) demonstrated that coumarin could inhibit the mutagenicity of 1-nitro-pyrene and 3nitrofluoranthene. Other studies have shown that coumarin can bind to DNA and interact with excision repair processes (Grigg, 1977). Genotoxicity studies have also been conducted on o-HPAA and 7-HC (i.e. the major metabolites of coumarin in the rat and human, respectively). Neither o-HPAA nor 7-HC at concentrations up to 5000 mg/plate showed any evidence of mutagenicity in S. typhimurium strains TA98, TA100, TA1535, TA1537 and TA1538 either with or without metabolic activation (Microbiological Associates, 1994a). Similarly, neither o-HPAA nor 7-HC at concentrations of up to 150 and 50 mg/mL, respectively, induced unscheduled DNA synthesis in cultured rat hepatocytes (Microbiological Associates, 1994b). Higher concentrations (.::;5000 llg/mL) of o-HPAA and
342
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
7-HC were cytotoxic to rat hepatocytes. In another study, o-HPAA at concentrations of up to 5000 mg/mL was reported not to produce chromosomal aberrations in CHO cells both with and without metabolic activation (Microbiological Associates, 1993a). However, a weak positive response was obtained in this test with 7-HC, but only at high, toxic concentrations and in the absence of metabolic activation (Microbiological Associates, 1993b).
Toxicity and Carcinogenicity Studies A number of studies have examined the acute, chronic and carcinogenic effects of coumarin in the rat and mouse. In the rat, single oral or ip doses (e.g. 125-500 mglkg) of coumarin result in a rapid depletion of hepatic nonprotein sulfYdryl groups [primarily reduced glutathione (GSH)], and after 24 h centrilobular hepatic necrosis and an elevation of plasma alanine and aspartate aminotransferase activities (Fentem et al., 1992a; Lake, 1984; Lake & Evans, 1993; Lake et al., 1989a, 1994a). Depending on the dose administered, coumarin treatment results in an increase in relative liver weight and changes in various hepatic biochemical parameters. Levels of total microsomal CYP and associated mixed-function oxidase enzymes tend to be reduced by coumarin administration especially after short periods of treatment. Microsomal glucose-6-phosphatase activity is also reduced. In contrast, coumarin administration leads to a sustained increase in nonprotein sulfydryl groups and certain enzyme activities such as GSH Stransferase and gamma-glutamyltransferase. Coumarin-induced lysosomal changes in rat liver have been demonstrated by histochemical and ultrastructural techniques (Grasso et al., 1974). Morphological examination of liver sections from coumarin-treated rats reveals a variety of changes, which are dependent on the magnitude of the dose and the duration of treatment. Such changes include necrosis, apoptosis, vacuolation, fatty change and bile duct hyperplasia. An intriguing aspect of coumarin-induced hepatotoxicity in the rat is the shift in the site of liver damage from the centrilobular to the periportal region after acute and chronic administration, respectively. Coumarin has been reported to be hepatotoxic and nephrotoxic in the dog at an oral dose of 100 mglkg given for periods of 8-22 days (Hagan et al., 1967; Hazleton et al., 1956). Morphological examination of liver sections revealed various lesions including cytoplasmic vacuolation, fatty change and necrosis. Hepatotoxic effects were also observed in dogs given 25 and 50 mglkg coumarin for longer periods, but not in dogs given 10 mglkg coumarin for up to 350 days (Hagan et al., 1967). A number of studies have examined the carcinogenicity of coumarin in the rat (Carlton et al., 1996; Griepentrog, 1973; Hagan et al., 1967; NTP, 1993a). Griepentrog (1973) reported that coumarin could produce bile duct carcinoma (cholangiocarcinoma) in rats given 0.5 and 0.6% coumarin, but
Metabolism and Pharmacokinetic Studies of Coumarins
343
not in animals receiving 0.1% and 0.25% coumarin. Species differences in coumarin-induced toxicity in vitro have been investigated in cultured hepatocytes (Ratanasavanh et al., 1996) and precision-cut liver slices (Price et al., 1996). The treatment of male Sprague-Dawley rat hepatocytes with 0.1, 0.25 and 0.5 mm coumarin resulted in toxicity as demonstrated by morphological examination of the cultures and by lactate dehydrogenase leakage (Ratanasavanh et al., 1996). Toxicity was also observed in hepatocyte cultures from male DBN2J strain mice and rabbits, whereas with cultured human hepatocytes toxicity was only observed with 1 mm coumarin and not with coumarin concentrations of 0.5 mm or below. Coumarin has also been reported to produce toxicity in cultured gerbil hepatocytes (Fentem et al., 1991b). Price et al. (1996) compared the toxicity of 0.5, 1 and 2 mm coumarin in 24-h cultured precision-cut male SpragueDawley rat, Dunkin-Hartley guinea pig, cynomolgus monkey and human liver slices. Based on the measurement of liver slice protein synthesis and potassium content coumarin produced concentration-dependent toxic effects in rat and guinea pig liver slices, whereas cynomolgus monkey and human liver slices were relatively resistant. These studies provide evidence for species differences in coumarininduced toxicity in vitro. The relative resistance of human and cynomolgus monkey liver slices and/or hepatocytes to coumarin toxicity correlates with coumarin 7-hydroxylation, the major pathway of coumarin metabolism in these species, being a detoxification pathway of coumarin metabolism.
Clinical Studies Coumarin is being evaluated for the treatment of various clinical conditions, resulting in the employment of a variety of dosing regimens. Recommended doses range from 8 mg for the treatment of venous constriction to 7000 mg! day in antineoplastic therapies (Marshall et al., 1994). While various mild side-effects have been reported following coumarin treatment, alterations to liver function have been noted in only a small proportion of patients receiving coumarin and tend to revert to normal after cessation of treatment. Reports of overt hepatotoxicity are rare (Cox et al., 1989). In a study with 45 renal cell carcinoma patients receiving 100 mg coumarin daily in combination with cimetidine, Marshall et al. (1987b) found no evidence for liver toxicity in any patient. In other trials with similar dosing regimens, groups of22 (Marshall et al., 1989), 24 (Marshall et al., 1987a), 50 (Dexeus et al., 1990) and 17 (Nolte et al., 1987) cancer patients showed no evidence of liver toxicity. Isolated cases of hepatotoxicity have been noted in other studies with patients receiving coumarin. Thus, Beinssen (1994) reported one possible case of hepatotoxicity due to coumarin treatment, and in another study Loprinzi et al. (1997) reported six cases of hepatotoxicity. Faurschou (1982) reported a case of toxic hepatitis in a patient given coumarin for 8 weeks, characterized by hepatomegaly and elevated serum
344
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
enzymes. All signs of liver toxicity returned to normal on cessation of treatment. It is clear from the published clinical studies that there is no clear relationship between coumarin dose and hepatotoxicity. For some individuals, bio- chemical signs of hepatotoxicity, such as elevated serum transaminases, return to normal despite continued treatment and in others return to normal after cessation of treatment. It is possible that the polymorphism in the human population with respect to CYP2A6 enzyme activity may account for the differential effects seen. However, no link between coumarin-induced hepatotoxicity and 7-hydroxylation status has yet been established.
Assessment of Risk to Humans The European Union Scientific Committee for Food has published an opinion on the levels of coumarin in natural flavouring source materials (Scientific Committee for Food, 1997). The limits for coumarin were less than 2 mg/kg in food and beverages with specific exceptions of 10 mg/kg in "special" caramels and in alcoholic beverages. While the Committee considered that further research on coumarin is desirable, it has recommended that the general limit for coumarin in food and beverages (due to the presence of natural source materials containing coumarin) should be lowered to the currently achievable limit of detection of 0.5 mg/kg. In order to assess the safety of coumarin from dietary sources and from fragrance use in cosmetic products for human health, it is necessary to consider the likely human exposure and in view of the marked species differences in coumarin metabolism and toxicity, the relevance ofthe rodent toxicity data to humans. Based on food intake data, with coumarin containing foods only accounting for 5% of total solid foodstuffs, the maximum daily intake for a 60-kg consumer would be 1.235 mg coumarin! day or 0.02 mg/kg/day. In addition, from fragrance use in cosmetic products the realistic daily exposure would be 2.289 mg/kg/day or 0.04 mg/kg/day for a 60-kg consumer. Both of these figs assume that all coumarin would be absorbed after either oral or dermal exposure and would be available to the systemic circulation. The total daily human coumarin exposure from the diet and from fragrance use in cosmetic products would thus be 0.06 mg/kg/day. This exposure level is over 2000 and over 3000 times lower, respectively, than those which produce liver tumours in rats (Carlton et al., 1996) and lung tumours in mice (NTP, 1993a). Unlike the rat and mouse, where high doses of coumarin can produce toxicity and carcinogenicity, there is little evidence of coumarin-induced toxicity in humans given therapeutic doses of coumarin which are up to 1900 times higher than those obtained from dietary sources and from fragrance use in cosmetic products. Such species differences are attributable
Metabolism and Pharmacokinetic Studies of Coumarins
345
to differences in coumarin biotransformation and disposition. Unlike the rat and mouse, where the 3,4-epoxidation pathway predominates, the major pathway of coumarin metabolism in humans is by 7-hydroxylation. Studies in mouse strains and the Mongolian gerbil have demonstrated that the 7hydroxylation pathway is a detoxification pathway of coumarin metabolism. However, while coumarin 7-hydroxylation is a detoxification pathway, this does not appear to be the only explanation for resistance of a species to coumarin-induced toxicity. For example, as demonstrated by in vitro (Lake et al., 1992a; Pearce et al., 1992) and limited in vivo (Gangolli et al., 1974; Lake et al., 1990) studies, the Syrian hamster is a poor 7-hydroxylator of coumarin, yet is resistant to chronic coumarin-induced liver injury. Unlike the rat, where large amounts of a coumarin dose appear in the faeces, species such as the Syrian hamster, baboon and humans excrete coumarin metabolites primarily in the urine. Such data suggest that the rat is an inappropriate animal model for the evaluation of the safety of coumarin for humans. Overall, 7-hydroxylation status is not the only factor responsible for determining the susceptibility of a species to coumarin-induced toxicity. While high 7-hydroxylation status appears to confer resistance to coumarininduced toxicity, some poor 7- hydroxylators are also not susceptible. The rat, mouse (i.e. B6C3F1 and CD-1 strains) and Syrian hamster are all poor 7-hydroxylators of coumarin. While the rat is susceptible to coumarininduced toxicity, the Syrian hamster is resistant and toxicity was only observed in a chronic gavage study in the mouse and not after dietary administration. Further studies on the metabolism and disposition of coumarin in these species may help clarify the precise mechanisms of coumarin -induced toxicity. A number of studies have examined the mutagenic and genotoxic potential of coumarin. Overall, the data suggest that coumarin is not a genotoxic agent. Indeed, the non-linear dose-response relationships for coumarin-induced tumour formation in rodents are typical of those of a non-genotoxic, rather than a genotoxic, carcinogen (Weisburger, 1994). As coumarin does not appear to be a genotoxic carcinogen in rodents, other mechanisms are required to explain why high doses of this compound can produce liver and lung tumours in some chronic studies. From the available metabolism data, coumarin is metabolized by the rat and mouse strains by the 3,4-epoxidation pathway to yield potentially toxic metabolites. In vitro studies have demonstrated that o-HPA, the major metabolite of coumarin in the rat and mouse, is more toxic to rat hepatocytes than the parent compound and a number of coumarin metabolites (Born et al., 1998b; Lake et al., 1989a). Certainly various aldehydes are known to produce toxic effects in biological systems (Feron et al., 1991). For example, formaldehyde carcinogenesis is a high-dose phenomenon in which cytotoxicity plays a crucial role (Feron et al., 1991). Additional studies are required to identify which metabolite(s) are responsible for coumarin-induced toxicity in rodents.
346
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Previous studies have demonstrated that while the majority of humans metabolize coumarin extensively to 7-HC, a deficiency in this pathway has been observed in some individuals which appears to be related to a genetic polymorphism in CYP2A6. The limited in vivo (Hadidi et al., 1997) and in vitro (van Iersel et al., 1994a) data available suggest that such deficient individuals will metabolize coumarin by the 3,4-epoxidation and possibly other pathways leading to the formation of 0- HPAA. In clinical studies occasional but reversible changes in biochemical markers of liver function have been observed in response to coumarin therapy. However, no relationship between coumarin 7-hydroxylation status and toxicity has been demonstrated in humans. In any event, considering the no-observedeffect levels (NOELs) observed in studies with experimental animals, the low level of exposure to coumarin from the diet and from fragrance use in cosmetic products would not be expected to produce any hepatotoxicity even in individuals with deficient coumarin 7-hydroxylase activity.
CONCLUSIONS Thus, by examination of available literature data, it can be concluded that coumarin exhibits marked species differences in both metabolism and toxicity and is not a genotoxic agent. Coumarin-induced hepatotoxicity has been found in the rat, mouse and dog, but not in humans may be due to predominance of 7-hydroxylation detoxification pathway. However, 7hydroxylation status is not the only determinant of susceptibility of a species to coumarin-induced toxicity. Evidence of liver and lung carcinogenicity in rat and mouse is probably attributable to non-genotoxic mechanisms, which only operate when there is target organ damage. Also, comparison of the NOELs for coumarin-induced toxicity and carcinogenecity with estimated human exposure from the diet and cosmetic products provides acceptable safety margins, such that coumarin exposure from these sources should pose no health risk to humans.
REFERENCES Beamand, J.A., Barton, P.T., Price, RJ. and Lake, B.G. (1998). Lack of effect of coumarin on unscheduled DNA synthesis in precision-cut human liver slices. Food and Chemical Toxicology, 36: 647-653. Behera, D., Damre, A., Varghese, A. and Veeranjaneyulu, A. (2008). In vitro evaluation of hepatic and extra-hepatic metabolism of coumarins using rat subcellular fractions: Correlation of in vitro clearance with in vivo data. Drug Metabolism and Drug Interactions, Communicated March 2008. Beinssen, A.P.A. (1994). Possible coumarin hepatotoxicity (Letter). Medical Journal of Australia, 161: 725. Bogan, D.P., Deasy, B., O'Kennedy, R and Smyth, M.R (1996). Interspecies differences in coumarin metabolism in liver micro somes examined by capillary electrophoresis. Xenobiotica, 26: 437-445. Booth, A.N., Masri, M.S., Robbins, D.J., Emerson, O.H., Jones, F.T. and DeEds, F. (1959). Urinary metabolites of coumarin and o-coumaric acid. Journal of Biological Chemistry, 234: 946-948.
Metabolism and Pharmacokinetic Studies of Coumarins
347
Born, S.L., Hu, J.K and Lehman-McKeeman, L.D. (1998b). o-Hydroxyphenylacetaldehyde (o-HPA) is a hepatotoxic metabolite of coumarin. Toxicological Sciences, 42: 400. Brown, D.L., Endean, D., Bodmer, S. and Cerveny, C. (1981). Effects of antioxidants on the mutagenicity of benzo(a)pyrene and 7,12-dimethylbenzanthracene. American Society for Microbiology, 81: 130. Bullock, P., Pearce, R., Draper, A., Podval, J., Bracken, W., Veltman, J., Thomas, P. and Parkinson, A. (1998). Induction of liver microsomal cytochrome P450 in Cynomolgus monkeys. Drug Metabolism and Disposition, 23: 736-748. Carlton, B.D., Aubrun, J.C. and Simon, G.S. (1996). Effects of coumarin following perinatal and chronic exposure in Sprague-Dawley rats and CD-1 mice. Fundamental and Applied Toxicology, 30: 145-151. Chang, T.KH. and Waxman, D.J. (1996). The CYF2A subfamily. In: Cytochromes P450: Metabolic and Toxicological Aspects, Ed. By C. Ioannides, CRC Press, Boca Raton, USA, pp.99-134. Cohen, A.J. (1979). Critical review of the toxicology of coumarin with special reference to interspecies differences in metabolism and hepatotoxic response and their significance to man. Food and Chemical Toxicology, 17: 277-289. Cox, D., O'Kennedy, R and Thomes, RD. (1989). The rarity ofliver toxicity in patients treated with coumarin (1,2-benzopyrone). Human and Experimental Toxicology, 8: 501-506. Creaven, P.J., Parke, D.V. and Williams, RT. (1965). A spectrofluorimetic study of the 7hydroxylation of coumarin by liver microsomes. Biochemical Journal, 96: 390-398. Dalet-Beluche, I., Boulenc, X., Fabre, G., Maurel, P. and Bonfils, C. (1992). Purification of two cytochrome P450 isozymes related to CYF2A and CYF3A gene families from monkey (baboon, Papio papio) liver microsomes. Cross reactivity with human forms. European Journal of Biochemistry, 204: 641-648. DeGarmo, O. and Raizman, P. (1967). Coumarin. In: Encyclopedia of Chemical Technology, Vol. 12, Ed. By Kirk, R.E. and D. F. Othmer, John Wiley & Sons, New York, pp. 425-433. den Besten, C., Korosi, S.A., Beamand, J.A., Walters, D.G. and Lake, B.G. (1990). Studies on the mechanism of coumarin-induced toxicity in rat hepatocytes. Toncology in Vitro, 4: 518-521. Dexeus, F.H., Logothetis, C.J., Sella, A., Fitz, K, Amato, R, Reuben, J.M. and Dozier, N. (1990). Phase II study of coumarin and cimetidine in patients with metastatic renal cell carcinoma. Journal of Clinical Oncology, 8: 325-329. Ding, S., Lake, B.G., Friedberg, T. and Wolf, C.R (1995). Expression and alternative splicing of the cytochrome P-450 CYF2A7. Biochemical Journal, 306: 161-166. Dominguez, K, Fentem, J.H., Garle, M.J. and Fry, J.R (1990). Comparison of Mongolian gerbil and rat hepatic microsomal mono-oxygenase activities: high coumarin 7-hydroxylase activity in the gerbil. Biochemical Pharmacology, 39: 1629-1631. Donato, M.T., Castell, J.V. and GOmez-Lech6n, M.J. (1998). The coumarin 7-hydroxylation assay in living hepatic cells in culture. ATLA, 26: 213-223. Edenharder, R and Tang, X. (1997). Inhibition of the mutagenicity of 2-nitrofluorene, 3nitrofluoranthene and 1-nitropyrene by flavonoids, coumarins, quinones and other phenolic compounds. Food and Chemical Toxicology, 35: 357-372. Egan, D., O'Kennedy, R, Moran, E., Cox, D., Prosser, E. and Thomes, RD. (1990). The pharmacology, metabolism, analysis, and applications of coumarin and coumarin-related compounds. Drug Metabolism Reviews, 22: 503-529. Faurschou, P. (1982). Toxic hepatitis due to benzo-pyrone. Human Toxicology, 1: 149-150. Fentem, J.H. and Fry, J.R (1991). Comparison of the effects of inducers of cytochrome P450 on Mongolian gerbil and rat hepatic microsomal monooxygenase activities. Xenobiotica, 21: 895-904. Fentem, J.H. and Fry, J.R (1992). Metabolism of coumarin by rat, gerbil and human liver microsomes. Xenobiotica, 22: 357-367. Fentem, J.H. and Fry, J.R (1993). Species differences in the metabolism and hepatotoxicity of coumarin. Comparitive Biochemistry and Physiology, 104C: 1-8. Fentem, J.H., Fry, J.R and Thomas, N.W. (1992a). Species differences in the hepatotoxicity of coumarin: a comparison of rat and Mongolian gerbil. Toxicology, 71: 129-136.
348
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Fentem, J.H., Fry, J.R. and Whiting, D.H. (1991a). o-Hydroxyphenylacetaldehyde : a major novel metabolite of coumarin formed by rat, gerbil and human liver microsomes. Bwchemical and Biophysical Research Communications, 179: 197-203. Fentem, J.H., Hammond, A.H. and Fry, J.R. (1991b). Maintenance of monooxygenase activities and induction of cytochrome P-450-mediated cytotoxicity in Mongolian gerbil hepatocyte cultures. Xenobiot~ca, 21: 1363-1370. Fernandez-Salguero, P., Hoffman, S.M.G., Cholerton, S., Mohrenweiser, H., Raunio, H., Rautio, A., Pelkonen, 0., Huang, J-D., Evans, W.E., Idle, J.R. and Gonzalez, F.J. (1995). A genetic polymorphism in coumarin 7-hydroxylation: sequence of the human CYF2A genes and identification of variant CYF2A6 alleles. Americal Journal of Human Genetics, 57: 651-660. Feron, V.J., Til, H.P., de Vrijer, F., Wouteresen, R.A., Cassee, F.R. and van Bladeren, P.J. (1991). Aldehydes: occurrence, carcinogenic potential, mechanism of action and risk assessment. Mutation Research, 259: 363-385. Feuer, G. (1970). 3-Hydroxylation of coumarin or 4-methylcoumarin by rat-liver microsomes and its induction by 4-methylcoumarin given orally. Chemico-Biological Interactions, 2: 203-216. Feuer, G. (1974). The metabolism and biological actions of coumarins. In: Progress in Medicinal Chemistry, Vol. 10, Ed. By Ellis, G.P. and G. B. West, North-Holland Publishing Company, Amsterdam, pp. 85-158. Forrester, L.M., Henderson, C.J., Glancey, M.J., Back, D.J., Park, B.K., Ball, S.E., Kitteringham, N.R., McLaren, A.W., Miles, J.L., Skett, P. and Wolf, C.R. (1992). Relative expression of cytochrome P450 isoenzymes in human liver and association with the metabolism of drugs and xenobiotics. Biochemical Journal, 281: 359-368. Galloway, S.M., Armstrong, M.J., Reuben, C., Colman, S., Brown, B., Cannon, C., Bloom, A.D., Nakamura, F., Ahmed, M., Duk, S., Rimpo, J., Margolin, B.H., Resnick, M.A. Anderson, B. and Zeiger, E. (1987). Chromosome aberrations and sister chromatid exchanges in Chinese hamster ovary cells: evaluations of 108 chemicals. Environmental and Molecular Mutagenesis, 10: 1-175. Gangolli, S.D., Shilling, W.H., Grasso, P. and Gaunt, I.F. (1974). Studies on the metabolism and hepatotoxicity of coumarin in the baboon. Biochem~cal Society Transactions, 2: 310-312. Gibbs, P.A., Janakidevi, K and Feuer, G. (1971). Metabolism of coumarin and 4-methylcoumarin by rat liver microsomes. Canadian Journal of Bwchem~stry, 49: 177-184. Grasso, P., Wright, M.G., Gangolli, S.D. and Hendy, R.J. (1974). Liver response tests. IX. Cytopathological changes in the enlarged but histologically normal rat liver. Food and Cosmetics Toxicology, 12: 341-350. Griepentrog, F. (1973). Pathologisch-anatomische befunde zur karzinogenen wirkung von cumarin im tierversuch. Toxicology, 1: 93-102. Grigg, G.W. (1977). Genetic effects of coumarins. Mutation Research, 47: 161-18l. Gullsten, H., Aglindez, J.A.G., Benitez, J., Laara, E., Ladero, J.M., Diaz-Rubio, M., FernandezSalguero, P., Gonzalez. F., Rautio, A., Pelkonen, O. and Raunio, H. (1997). CYF2A6 gene . polymorphism and risk of liver cancer and cirrhosis. Pharmacogenetics, 7: 247-250. Hadidi, H., Zahlsen, K, Idle, J.R. and Cholerton, S. (1997). A single amino acid substitution (Leu160His) in cytochrome P450 CYF2A6 causes switching from 7-hydroxylation to 3hydroxylation of coumarin. Food and Chem~cal Toxicology, 35: 903-907. Hagan, E.C., Hansen, W.H., Fitzhugh, O.G., Jenner, P.M., Jones, W.I., Taylor, J.M., Long, E.L., Nelson, A.A. and Brouwer, J.B. (1967). Food flavourings and compounds of related structure. II. Subacute and chronic toxicity. Food and Cosmetics Toxicology, 5: 141-157. Hardt, T.J. and Ritschel, W.A. (1983). Dose-related pharmacokinetics of coumarin, 7hydroxycoumarin and 7 -hydroxycoumarin glucuronide upon intraperitoneal administration of coumarin and 7-hydroxycoumarin in the rat. Arzneimittel-Forschung (Drug Research), 33: 1442-1446. Hawley, G. (1971). The Condensed Chemical Dictionary, Van Nostrand Reinhold, New York. Hazleton, L.W., Tusing, T.W., Zeitlin, B.R., Thiessen, R., Jr. and Murer, H.K. (1956). Toxicity of coumarin. Journal of Pharmacology and Experimental Therapeutics, 118: 348-358.
Metabolism and Pharmacokinetic Studies of Coumarins
349
Honkakoski, P. and Negishi, M. (1997). The structure, function, and regulation of cytochrome P450 2A enzymes. Drug Metabolism Reviews, 29: 977-996. Huntingdon Life Sciences (1996a). 14C-Coumarin. Dermal absorption in man. Project Number RIF 30/943257. Report to RIFM. Ide, F., Ishikawa, T. and Takayama, S. (1981). Detection of chemical carcinogens by assay of unscheduled DNA synthesis in rat tracheal epithelium in short-term organ culture. Journal of Cancer Research and Clinical Oncology, 102: 115-126. Imanishi, H., Sasaki, Y.F., Matsumoto, K, Watanabe, M., Ohta, T., Shirasu, Y. and Tutikawa, K (1990). Suppression of 6-TG-resistant mutations in V79 cells and recessive spot formations in mice by vanillin. Mutation Research, 243: 151-158. Kaighen, M. and Williams, R.T. (1961). The metabolism of [3- 14 Clcoumarin. Journal of Medicinal Chem£stry, 3: 25-43. Kratz, F. (1976). Coumarin 7-hydroxylase activity in microsomes from needle biopsies of normal and diseased human liver. European Journal of Clinical Pharmacology, 10: 131137. Lake, B.G. (1984). Investigations into the mechanism of coumarin-induced hepatotoxicity in the rat. Archwes of Toxicology, 7: 16-29. Lake, B.G. and Evans, J.G. (1993). Effect of pretreatment with some mixed-function oxidase enzyme inducers on the acute hepatotoxicity of coumarin in the rat. Food and Chemical Toxicology, 31: 963-970. Lake, B.G., Evans, J.G., Lewis, D.F.V. and Price, R.J. (1994a). Studies on the acute effects of coumarin and some coumarin derivatives in the rat. Food and Chemical Toxicology, 32: 357-363. Lake, B.G., Evans, J.G., Walters, D.G. and Price, R.J. (1990), Studies on the hepatotoxicity and metabolism of coumarin in the rat and Syrian hamster. Human and Experimental Toxicology, 9: 356. Lake, B.G., Gaudin, H., Price, R.J. and Walters, D.G. (1992a). Metabolism of [3- 14Clcoumarin to polar and covalently bound products by hepatic microsomes from the rat, Syrian hamster, gerbil and humans. Food and Chemical Toxicology, 30: 105-115. Lake, B.G., Gray, T.J.B., Evans, J.G., Lewis, D.F.v., Beamand, J.A. and Hue, KL. (1989a). Studies on the mechanism of coumarin-induced toxicity in rat hepatocytes: comparison with dihydrocoumarin and other coumarin metabolites. Toxicology and Apphed Pharmacology, 97: 311-323. Lake, B.G., Osborne, D.J., Walters, D.G. and Price, R.J. (1992b). Indentification of 0hydroxyphenylacetaldehyde as a major metabolite of coumarin in rat hepatic microsomes. Food and Chem£cal Toxicology, 30: 99-104. Lake, B.G., Sauer, M.J., Esclangon, F., Beamand, J.A., Price, R.J. and Walters, D.G. (1995). Metabolism of coumarin by precision-cut calf liver slices and calf liver microsomes. Xenobiotica, 25: 133-141. Lake, B.G., Walters, D.G. and Gangolli, S.D. (1989b). Comparison of the metabolism and disposition of [3- 14Clcoumarin in the rat and marmoset (Callithnx jacchus). Toxicology Letters, 45: 299-306. Loprinzi, C.L., Sloan, J. and Kugler, J. (1997). Coumarin-induced hepatotoxicity. Journal of Clinical Oncology, 15: 3167-3168. Lovell, D.P., van Iersel, M., Walters, D.G., Price, R.J. and Lake, B.G. (1999). Genetic variation in the metabolism of coumarin in mouse liver. Pharmacogenetics, 9: 239-250. Lush, I.E. and Andrews, KM. (1978). Genetic variation between mice in their metabolism of coumarin and its derivatives. Genetics Research, 31: 177-186. Marshall, M.E., Butler, K, Cantrell, J., Wiseman, C. and Mendelsohn, L. (1989). Treatment of advanced malignant melanoma with coumarin and cimetidine: a pilot study. Cancer Chemotherapy and Pharmacology, 24: 65-66. Marshall, M.E., Mendelsohn, L., Butler, K, Cantrell, J., Harvey, J. and Macdonald, J. (1987a). Treatment of non-small cell lung cancer with coumarin and cimetidine. Cancer Treatment Reports, 71: 91-92. Marshall, M.E., Mendelsohn, L., Butler, K, Riley, L., Cantrell, J., Wiseman, C. Taylor, R. and Macdonald, J.S. (1987b). Treatment of metastatic renal cell carcinoma with
350
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
coumarin (1,2-benzopyrone) and cimetidine: a pilot study. Journal of Clinical Oncology, 5: 862-866. Marshall, M.E., Mohler, J.L., Edmonds, K., Williams, B., Butler, K., Ryles, M., Weiss, L., Urban, D., Bueschen, A., Markiewicz, M. and Cloud, G. (1994). An updated review of the clinical development of coumarin (1,2-benzopyrone) and 7-hydroxycoumarin. Journal of Cancer Research and Clinical Oncology, 120: S39-S42. Maurice, M., Emiliani, S., Dalet-Beluche, I., Derancourt, J. and Lange, R (1991). Isolation and characterization of a cytochrome P450 of the IIA subfamily from human liver microsomes. European Journal of Biochemistry, 200: 511-517. Mead, J.A.R., Smith, J.N. and Williams, RT. (1958a). Studies in detoxication. 72. The metabolism of coumarin and of o-coumaric acid. Biochemical Journal, 68: 67-74. Mead, J.A.R., Smith, J.N. and Williams, RT. (1958b). Studies in detoxication. 71. The metabolism of hydroxycoumarins. Biochemical Journal, 68: 61-67. Microbiological Associates (1993a). Chromosome aberrations in Chinese hamster ovary (CHO) cells. Laboratory Study Number TC91O. 337. Report to RIFM. Microbiological Associates (1993b). Chromosome aberrations in Chinese hamster ovary (CHO) cells. Laboratory Study Number TC911. 337. Report to RIFM. Microbiological Associates (1994a). Salmonella/mammalian-microsome plate incorporation mutagenicity assay (Ames test) Laboratory Study Numbers TC910. 501 and TC911. 501. Report to RIFM. Microbiological Associates (1994b). Unscheduled DNA synthesis in rat primary hepatocytes. Laboratory Study Numbers TC910. 380 and TC911. 380. Report to RIFM. Miles, J.S., McLaren, A.W., Forrester, L.M., Glancey, M.J., Lang, M.A. and Wolf, C.R (1990). Identification of the human liver cytochrome P-450 responsible for coumarin 7-hydroxylase activity. Biochemical Journal, 267: 365-371. Morris, D.L. and Ward, J.B., Jr. (1992). Coumarin inhibits micronuclei formation induced by benzo(a)pyrene in male but not female ICR mice. Environmental and Molecular Mutagenesis, 19: 132-138. National Toxicology Program (1993a). Toxicology and carcinogenesis studies of coumarin (CAS No. 91-64-5) in F344IN rats and B6C3F1 mice (gavage studies). Technical Report No. NTP TR 422. NIH Publication No. 93-3153, US Department of Health and Human Services, NIH, Research Triangle Park, NC. Negishi, M., Lindberg, R., Burkhart, B., Ichikawa, T., Honkakoski, P. and Lang, M. (1989). Mouse steroid 15a-hydroxylase gene family: identification of type II P-450 15a as coumarin 7-hydroxylase. Biochemistry, 28: 4169-4172. Nelson, D.R, Koymans, L., Kamataki, T., Stegeman, J.J., Feyereisen, R., Waxman, D.J., Waterman, M.R, Gotoh, 0., Coon, M.J., Estabrook, RW., Gunsalus, I.C. and Nebert, D.W. (1996). P450 superfamily: update on new sequences, gene mapping, accession numbers and nomenclature. Pharmacogenetics, 6: 1-42. Nolte, H., Pedersen, L. and Mouridsen, H.T. (1987). Combined treatment of advanced malignant melanoma with coumarin and cimetidine. Anticancer Research, 7: 449-450. Opdyke, D.L.J. (1974). Monographs on fragrance raw materials: coumarin. Food and Cosmetics Toxicology, 12: 385-388. Pasanen, M., Rannala, Z., Tooming, A., Sotaniemi, E.A., Pelkonen, O. and Rautio, A. (1997). Hepatitis A impairs the function of human hepatic CYF2A6 in vivo. Toxicology, 123: 177-184. Pearce, R, Greenway, D. and Parkinson, A. (1992). Species differences and interindividual variation in liver microsomal cytochrome P450 2A enzymes: effects on coumarin dicumarol, and testosterone oxidation. Archives of Biochemistry and Biophysics, 298: 211-225. Pelkonen, 0., Raunio, H., Rautio, A., Maenpaa, J. and Lang, M.A. (1993). Coumarin 7hydroxylase: characteristics and regulation in mouse and man. Journal of the Irish Colleges of Physicians and Surgeons, 22: 24-28. Pelkonen, 0., Raunio, H., Rautio, A., Pasanen, M. and Lang, M.A. (1997). The metabolism of coumarin. In: Coumarins: Biology, Applications and Mode of Action, Ed. By O'Kennedy R and RD. Thornes, John Wiley & Sons, USA, pp. 67-92.
Metabolism and Pharmacokinetic Studies of Coumarins
351
Pelkonen, 0., Sotaniemi, E.A. and Ahokas, J.T. (1985). Coumarin 7-hydroxylase activity in human liver microsomes. Properties of the enzyme and interspecies comparisons. British Journal of Clinical Pharmacology, 19: 59-66. Peters, M.M.C.G., Walters, D.G., van Ommen, B., van Bladeren, P.J. and Lake, B.G. (1991). Effect of inducers of cytochrome P-450 on the metabolism of [3- 14Clcoumarin by rat hepatic microsomes. Xenobiotica, 21: 499-514. Piller, N.B. (1977). (3_ 14C)Coumarin distribution in rat tissues after the injection of a single dose. Research in Experimental Medicine, 171: 93-100. Price, RJ., Mistry, H., Wield, P.T., Renwick, A.B., Beamand, J.A. and Lake, B.G. (1996). Comparison of the toxicity of allyl alcohol, coumarin and menadione in precision-cut rat, guinea-pig, Cynomolgus monkey and human liver slices. Archives of Toxicology, 71: 107111. Price, RJ., Renwick, A.B., Beamand, J.A., Esclangon, F., Wield, P.T., Walters, D.G. and Lake, B.G. (1995). Comparison of the metabolism of 7-ethoxycoumarin and coumarin in precision-cut rat liver and lung slices. Food and Chemical Toxicology, 33: 233-237. Ratanasavanh, D., Lamiable, D., Biour, M., Guedes, Y., Gersberg, M., Leutenegger, E. and Riche, C. (1996) Metabolism and toxicity of coumarin on cultured human, rat, mouse and rabbit hepatocytes. Fundamental and Chmcal Pharmacology, 10: 504-510. Raunio, H., Syngelma, T., Pasanen, M., Juvonen, R, Honkakoski, P., Kairaluoma, M.A., Sotaniemi, E., Lang, M.A. and Pelkonen, O. (1988b). Immunochemical and catalytical studies on hepatic coumarin 7-hydroxylase in man, rat, and mouse. Biochemical Pharmacology, 37: 3889-3895. Ritschel, W.A. and Grummich, KW. (1981). Pharmacokinetics of coumarin and 7hydroxycoumarin upon i. v, p. o. administration in the euthyroid and hypothyroid beagle dog. Arzneimittel-Forschung (Drug Research), 31: 643-649. Ritschel, W.A., Brady, M.E. and Tan, H.S.1. (1979). First-pass effect of coumarin in man. International Journal of Clinical Pharmacology and Biopharmacy, 17: 99-103. Ritschel, W.A., Brady, M.E., Tan, H.S.I., Hoffman, KA., Yiu, I.M. and Grummich, KW. (1977). Pharmacokinetics of coumarin and its 7-hydroxy-metabolites upon intravenous and peroral administration of coumarin in man. European Journal of Clinical Pharmacology, 12: 457-461. Ritschel, W.A., Denson, D.D. and Grummich, KW. (1988). Pharmacokinetics of coumarin and 7-hydroxycoumarin in the rhesus monkey after intravenous and peroral administration. Arzneimtttel-Forschung (Drug Research), 38: 1619-1623. Ritschel, W.A. and Hardt, T.J. (1983). Pharmacokinetics of coumarin, 7-hydroxycoumarin and 7-hydroxycoumarin glucuronide in the blood and brain of gerbils following intraperitoneal administration of coumarin. Arzneimittel-Forschung (Drug Research), 33: 1254-1258. Ritschel, W.A. and Hoffman, KA. (1981). Pilot study on bioavailability of coumarin and 7hydroxycoumarin upon peroral administration of coumarin in a sustained-release dosage form. Journal of Clinical Pharmacology, 21: 294-300. Ritschel, W.A., Hoffmann, KA., Tan, H.S. and Sanders, P.R (1976). Pharmacokinetics of coumarin upon i. v. administration in man. Arzneimittel-Forschung (Drug Research), 26: 1382-1387. Ritschel, W.A. and Hussain, S.A. (1988). Transdermal absorption and topical bioavailability of coumarin. Methods and Findings in Experimental and Clinical Pharmacology, 10: 165169. Sanyal, R, Darroudi, F., Parzefall, W., Nagao, M. and Knasmuller, S. (1997). Inhibition of the genotoxic effects of heterocyclic amines in human derived hepatoma cells by dietary bioantimutagens. Mutagenesis, 12: 297-303. Sasaki, Y.F., Imanishi, H., Ohta, T. and Shirasu, Y. (1987). Effects of antimutagenic flavourings on SCEs induced by chemical mutagens in cultured Chinese hamster cells. Mutation Research, 189: 313-318. Sasaki, Y.F., Imanishi, H., Watanabe, M., Ohta, T. and Shirasu, Y. (1990). Suppressing effect of antimutagenic flavorings on chromosome aberrations induced by UV-light or Xrays in cultured Chinese hamster cells. Mutation Research, 229: 1-10.
352
Comp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Scheline, RR (1968). Studies on the role of the intestinal microflora in the metabolism of coumarin in rats. Acta Pharmacologica et Toxicologica, 26: 325-331. Scientific Committee for Food (1997). Opinion on coumarin (a constituent of natural flavouring source materials limited by Annex II of Flavourings Directive 88/388IEEC). Expressed on 16 December 1994. Reports of the Scientific Committee for Food (Thirtysixth series). pp. 13-20. European Communities, 1997. Shilling, W.H., Crampton, RF. and Longland, RC. (1969). Metabolism of coumarin in man. Nature, 221: 664-665. Shimada, T., Yamazaki, H. and Guengerich, F.P. (1996). Ethnic-related differences in coumarin 7-hydroxylation activities catalyzed by cytochrome P4502A6 in liver microsomes of Japanese and Caucasian populations. Xenobiotica, 26: 395-403. Shimada, T., Yamazaki, H., Mimura, M., Inui, Y. and Guengerich, F.P. (1994). Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. Journal of Pharmacology and Experimental Therapeutics, 270: 414-423. Soine, T.O. (1964). Naturally occurring coumarins and related physiological activities. Journal of Pharmaceutical Sciences, 53: 231-264. Sotaniemi, E.A., Rautio, A., Backstrom, M., Arvela, P. and Pelkonen, O. (1995). CYP3A4 and CYP2A6 activities marked by the metabolism of lignocaine and coumarin in patients with liver and kidney diseases and epileptic patients. British Journal of Clinical Pharmacology, 39: 71-76. Squires, E.J. and Negishi, M. (1990). Tissue-specific regulation of cytochrome P-450 dependent testosterone 151l-hydroxylase. Canadian Journal of Physiology and Pharmacology, 68: 769-776. Steensma, A., Beamand, J.A., Walters, D.G., Price, RJ. and Lake, B.G. (1994). Metabolism of coumarin and 7-ethoxycoumarin by rat, mouse, guinea pig, Cynomolgus monkey and human precision-cut liver slices. Xenobiotica, 24: 893-907. TNO (1996). Volatile Compounds in Food, Qualitative and Quantitative Data, TNO, The Netherlands. Valencia, R, Mason, J.M. and Zimmering, S. (1989). Chemical mutagenesis testing in Drosophila. VI. Interlaboratory comparison of mutagenicity tests after treatment of larvae. Environmental and Molecular Mutagenesis, 14: 238-244. van Iersel, M.L.P.S., Henderson, C.J., Walters, D.G., Price, RJ., Wolf, C.R and Lake, B.G. (1994a). Metabolism of [3- 14Clcoumarin by human liver microsomes. Xenobiotica, 24: 795-803. van Iersel, M., Walters, D.G., Price, RJ., Lovell, D.P. and Lake, B.G. (1994b). Sex and strain differences in mouse hepatic coumarin 7-hydroxylase activity. Food and Chemical Toxicology, 32: 387-390. van Sumere, C.F. and Teuchy, H. (1971). The metabolism of [2_ 14 Clcoumarin and [2-14Cl7-hydroxycoumarin in the rat. Archives Internationales de Physiologie et de Biochimie, 79: 665-679. Waller, A.R and Chasseaud, L.F. (1981). The metabolic fate of [l4Clcoumarin in baboons. Food and Chemical Toxicology, 19: 1-6. Walters, D.G., Lake, B.G. and Cottrell, R.C. (1980). High-performance liquid chromatography of coumarin and its metabolites. Journal of Chromatography, 196: 501-505. Weisburger, J.H. (1994). Does the Delaney clause of the U.S. Food and Drug Laws prevent human cancers? Fundamental and Apphed Toxicology, 22: 483-493. Williams, R.T., Millburn, P. and Smith, R.L. (1965). The influence of enterohepatic circulation on toxicity of drugs. Annals of the New York Academy of Sciences, 123: 110-124. Wrighton, S.A. and Stevens, J.C. (1992). The human hepatic cytochromes P450 involved in drug metabolism. CRC Critical Reviews in Toxicology, 22: 1-21.
Metabolism and Pharmacokinetic Studies of Coumarins
353
Yamano, S., Tatsuno, J. and Gonzalez, F.J. (1990). The CYP2A3 gene product catalyzes coumarin 7-hydroxylation in human liver microsomes. Biochemistry, 29: 1322-1329. Yoon, J.S., Mason, J.M., Valencia, R., Woodruff, R.C. and Zimmering, S. (1985). Chemical mutagenesis testing in Drosophila. IV. Results of 45 coded compounds tested for the National Toxicology Program. EnVIronmental Mutagenes~s, 7: 349-367. Yun, C-H., Shimada, T. and Guengerich, F.P. (1991). Purification and characterization of human liver microsomal cytochrome P-450 2A6. Molecular Pharmacology, 40: 679-685.
"This page is Intentionally Left Blank"
18 N aringin and its Aglycone, Research and Managements RICKY W.K. WONG 1,* AND FANNY Y.F. YOUNG 2
ABSTRACT
The grapefruit extract components, naringin and its aglycone naringinin are commonly used as health supplements; they exert a variety of pharmacological actions. This article attempts to review their pharmacokinetics, pharmacological actions and their uses in various managements, including effect on cardiovascular system; effect on skeletal system; effect on smooth muscle; effect on gastric intestinal system; effect on endocrine system; effect against tumour; protection against toxins in chemotherapy drugs and the environment; antioxidant effect; drug interactions; antiinflammatory effect and the newly discovered osteogenic and antibacterial actions. Key words : N aringin, naringenin, antioxidant effect, drug interactions, osteogenic effect, antibacterial effect, anti-inflammatory effect
INTRODUCTION Naringin is a flavonoid compound found in grapefruit and other citrus fruit, which gives grapefruit its characteristic bitter flavor. Naringin is believed to enhance our perception of taste by stimulating the taste buds. N aringin and its aglycone naringinin, are commonly used as health supplements; they exert a variety of pharmacological actions. For example, 1. 2IF, Prince Philip Dental Hospital, 34 Hospital Road, Sai Ying Pun,Hong Kong
(University of Hong Kong). 2. Hong Kong Shue Yan University. * Corresponding author: E-mail: [email protected]
356
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
it may be instrumental in inhibiting cancer-causing compounds and thus may have potential chemotherapeutic value. On the other hand, studies have also shown that naringin interferes with enzymatic activity in the intestines and, thus, with the breakdown of certain drugs, resulting in higher blood levels of the drug. In order to summarize their actions on health, a MEDLINE search covering the period 1977-2007 was performed to identify review articles, studies, and case reports referencing the biological properties of naringin and naringenin. The aim/method, major results and conclusion/actions were tabulated in the following sections for easy reference: Pharmacokinetics Effects on cardiovascular system Effects on skeletal system Effects on smooth muscle Effects on gastric intestinal system Effects on endocrine system Effects against tumour Protections against toxins in chemotherapy drugs and the environment Antioxidant effects Antimicrobial effects Drug interactions Antiinflammatory effects Other effects
RESEARCH ON NARINGIN AND NARENGININ SHOWS THE FOLLOWINGS: Pharmacokinetics Action/Conclusion Absorbed citrus flavanones may undergo glucuronidation before urinary excretion (Ameer et al., 1996).
AimlMethod Disposition of citrus flavonoids was evaluated after single oral doses of pure compounds (500 mg naringin and 500 mg hesperidin) and after multiple doses of combined grapefruit juice and orange juice and of once-daily grapefruit.
Results Cumulative urinary recovery indicated low bioavailability «25%) of naringin and hesperidin. The aglycones naringenin and hesperidin were detected in urine and plasma.
Table Contd.
Naringin and its Aglycone, Research and Managements
357
Contd.
Action/Conclusion
AimlMethod
Extensive glucuronidationl sulfation of naringenin occurred during the first pass at gut wall (Hsiu et al., 2002).
N aringenin was administered intravenously and orally to rabbits, and naringin was administered orally. The concentration of naringenin in serum prior to and after enzymatic hydrolysis was determined by HPLC method.
Oral administration of the flavanone aglycones, hesperetin and naringenin, lead to their rapid absorption as their conjugated forms. The cumulative urinary recovery data indicated low bioavailability for both flavanone aglycones, owing to extensive first-pass metabolism partly by cleavage of the C-ring by the enzymes of intestinal bacteria leading to degradation products such as phenolic acids (Kanaze et al., 2007).
Six healthy volunteers received orally 135 mg of each compound, hesperetin and naringenin, under fasting conditions. Blood samples were collected at 14 different time points over a 12 h period. Urine was collected over 24 h, in five sequential timed intervals.
Results The absolute bioavailability of oral naringenin was only 4%, whereas after taking the conjugated naringenin into account, it increased to 8%. When naringin was administered orally, only little naringenin and predominantly its glucuronidesl sulfates were circulating in the plasma Pharmacokinetic analysis showed that both hesperetin and naringenin were rapidly absorbed and their concentrations in plasma observed 20 min after dosing and reached a peak in 4.0 and 3.5 h, respectively. The mean peak plasma concentration (C(max)) for hesperetin and naringenin were 825.78 + 1410.63 ng/mL (2731.8 + 11358.4 nmol/l) and 2009.51 + 1-770.82 ng/mL (7386.6 + 12833.4 nmol!ll, respectively and the mean AUC(O-infinity) values were 4846.20 + 11675.99 ng h/mL and 9424.52 + 1-2960.52 ng hlmL for hesperetin and naringenin, respectively. The elimination half-life for hesperetin was found to be 3.05+1 -0.91 h and for naringenin 2.31 + 1-0.40 h, respectively. The mean values of the relative cumulative urinary excretion, as percentage of the administered dose, for hesperetin and naringenin, were found to be 3.26 + 1-0.44 and 5.81 + 1-0.81%, respectively. Table Contd.
358
Camp. Bio. Nat. Prod., VoL 2 - Efficacy, Safety & Clinical Evaluation I
Contd.
Action/Conclusion The results ofthe present study demonstrate the absorption of grapefruit flavanones via the presence of their metabolites in plasma (Mata-Bilbao Mde et at., 2007).
AimlMethod Ten healthy beagles were administered 70 mg citrus flavonoids as a grapefruit extract contained in capsules, while two additional dogs were used as controls and given an excipient.
Results N aringin reached its maximun plasma concentration at around 80 min, whereas naringenin and naringenin glucuronide reached their maximun plasma concentrations at around 20 and 30 min, respectively. Maximum plasma concentrations of naringin, naringenin and naringenin glucuronide (medians and ranges) were 0.24 (0.05-2.08), 0.021 (0.001-0.3) and 0.09 (0.034-0.12) microm01l1, respectively. The areas under the curves were 23.16 I (14.04-70.62) min x micromollfor nariningin, 1.78 (0.09-4.95) min x microm01l1 for naringenin and 22.5 (2.7499.23) min x micromolll for naringenin glucuronide. The median and range values for mean residence time were 3.3 (1.5-9.3), 2.8 (0.8-11.2) and 8.0 (2.3-13.1) h for naringin, naringenin and naringenin glucuronide, respectively.
Effects on cardiovascular system Action/Conclusion Ingestion of grapefruit lowers elevated hematocrits in human subjects (Robbins et al., 1988).
AimlMethod The effect on hematocrits of adding grapefruit to the daily diet was determined using 36 human subjects (12 F, 24 M) over a 42-day study.
Results The hematocrits ranged from 36.5 to 55.8% at the start and 38.8% to 49.2% at the end of the study.
Table Contd.
Naringin and its Aglycone, Research and Managements
359
Contd.
Action/Conclusion
AimlMethod
Results
Plasma and hepatic cholesterol and hepatic activities of 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase and acyl CoA: cholesterol transferase (ACAT) are lower in rats fed citrus peel extract or a mixture of citrus bioflavonoids (Bok et al., 1999).
The cholesterol-lowering effects of tangerine peel extract and a mixture of two citrus flavonoids were tested.
The inhibition of HMG-CoA reductase and ACAT activities resulting from either tangerine-peel extract or its bioflavonoids could account for the decrease in fecal neutral sterol.
Hypocholesterolemic effect of naringin associated with hepatic cholesterol regulating enzyme changes in rats (Shin et al., 1999).
The effects of the citrus bioflavonoid naringin were tested by using it as a supplement in a highcholesterol diet.
Anti-atherogenic effect of citrus flavonoids, naringin and naringenin (Lee et al., 2001).
The anti-atherogenic effects of the citrus flavonoids, naringin and naringenin, were evaluated in high cholesterol-fed rabbits.
Interactive effect of naringin and vitamin E on cholesterol biosynthesis (Choi et al., 2001).
The interactive effect of naringin and vitamin E was studied with respect to cholesterol metabolism and antioxidant status in highcholesterol-fed rats.
N aringin has an anti atherogenic effect with the inhibition of intercellular adhesion molecule-1 in hypercholesterolemic rabbits (Choe et al., 2001).
This study evaluated the effect of naringin on blood lipid levels and aortic fatty streaks, and its action mechanism in hypercholesterolemic rabbits.
The combination of the inhibited HMG-CoA reductase (-24.4%) and ACAT (20.2%) activities as a result of naringin supplementation could account for the decrease offecal neutral sterols. The anti-atherogenic effect of the citrus flavonoids, naringin and naringenin, is involved with a decreased hepatic ACAT activity and with the downregulation of VCAM-1 and MCP-1 gene expression. N aringin lowers the plasma lipid concentrations when the dietary vitamin E level is low. The HMG-CoA reductase-inhibitory effect ofnaringin was more potent when dietary vitamin E was at a normal level. N aringin treatment inhibited hypercholesterolemiainduced intercellular adhesion molecule-1 (ICAM-1) expression on endothelial cells. Hypercholesterolemia caused fatty liver and elevation of liver enzymes, which was prevented by naringin but not by lovastatin. Table Con/d.
360
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Contd. Action/Conclusion
AimlMethod
Results
Hypocholesterolemic effect of naringin and rutin flavonoids (da Silva et al., 2001).
The effect of naringin and rutin on the metabolism lipidic of chicks hypercholesterolemic was evaluated.
Naringin and rutin reduced the levels of total cholesterol significantly, cholesterol-LDL, cholesterol-VLDL and triglycerols, not presenting, however, reductions in the levels of cholesterol-HDL.
Flavonoids prevent in vitro LDL oxidation and probably would be important to prevent atherosclerosis (Naderi et al., 2003).
The susceptibility of LDL to in vitro oxidation was assessed. LDL oxidation were monitored by change in 234absorbance in the presence and absence of pure flavonoids. The effect of naringin on hypercholesterolemic subjects was studied. A hypercholesterolemic group (n=30) and healthy control group (n=30) were established.
Genistein, morin and naringin have stronger inhibitory activity against LDL oxidation than biochanin A or apigenin.
N aringin may play an important role in lowering plasma cholesterol and regulating the antioxidant capacity in hypercholesterolemic subjects (Jung et al., 2003).
N aringin lowers the plasma cholesterol level via the inhibition of hepatic HMG-CoA reductase activity and improve the activities of hepatic antioxidant enzymes against oxidative stress (Kim et al., 2004).
The lipid lowering and antioxidant capacity of naringin was evaluated in LDL receptor knockout (LDLR-KO) mice fed a cholesterol (0.1 g/ 100 g) diet.
Flavonoids that showed strong increase of ocular blood flow also showed marked increase of retinal function recovery (Chiou & Xu, 2004).
Effects of flavonoids to improve retinal function recovery after ischemic insult were studied. Electroretinography was used to measure the b-wave recovery as an indication of retinal function recovery.
Naringin supplementation was found to lower the plasma total cholesterol by 14% and low-density lipoprotein cholesterol concentrations by 17%, apolipoprotein B levels were significantly lowered, erythrocyte superoxide dismutase and catalase activities were significantly increased. The hepatic HMG-CoA reductase activity was significantly lower in the naringin and lovastatin supplemented groups than in the control group, the superoxide dismutase, catalase, and glutathione reductase activities were all significantly higher in the naringin-supplemented group than in the control group. Naringenin, hesperetin, and rutin were found to produce marked positive effects on bwave recovery, whereas naringin, hesperidin, and quercetin showed poor recovery of b-wave after ischemic insult of the retina. Table Contd.
361
Naringin and its Aglycone, Research and Managements Contd.
Action/Conclusion
AimlMethod
Results
Reactive oxygen species (ROS) playa causal role in renal ischemia/ reperfusion (I/R) induced renal injury and naringin exert renoprotective effects probably by the radical scavenging and antioxidant activities (Singh & Chopra, 2004). Both naringin and lovastatin contributed to hypocholesterolemic action. Naringin seemed to preserve tissue morphology from damages induced by high cholesterol diet (Jeon et al., 2004). The vasorelaxant effects of (+/-)-naringenin seem to be basically related to the inhibition of phosphodiesterase (PDE)1, PDE4 and PDE5 activities (OralIo et al., 2005).
The protective effect of naringin against the damage inflicted by ROS during renal I1R was investigated in Sprague-Dawley rats using histopathological and biochemical parameters.
Pretreatment of animals with naringin markedly attenuated renal dysfunction, morphological alterations, reduced elevated TBARS levels and restored the depleted renal antioxidant enzymes.
To confirm the hypocholesterolemic role of naringin, male rabbits were fed 0.5% high-cholesterol diet or high-cholesterol diet supplemented with either 0.05% naringin or 0.03% lovastatin for 8 weeks.
The naringin and lovastatin supplements significantly lowered plasma total- and LDLcholesterol and hepatic lipids levels, while significantly increasing HDL-C/total-C ratio compared to the control group.
The potential vasorelaxant, antioxidant and cyclic nucleotide PDE inhibitory effects of the citrus-fruit flavonoids naringin and (+/-)-naringenin were comparatively studied.
(+/-)-naringenin relaxed, in a concentration-dependent manner, the contractions elicited by phenylephrine (PHE, 1 microM) or by a high extracellular KCI concentration (60 mM) in intact rat aortic rings.
The active ingredients associated with the antihypertensive effect of sweetie juice are the flavonoids naringin and narirutin (Reshef et al., 2005).
Patients with stage I hypertension, the antihypertensive effect of juice of the so-called sweetie fruit (a hybrid between grapefruit and pummelo) with and without high flavonoid content were studied. The effect of the flavonoids hesperidin and naringin on glucose and lipid regulation in C57BL/KsJ-db/db mice was studied.
The high-flavonoid (HF) sweetie juice was more effective than LF sweetie juice in reducing diastolic blood pressure.
Hesperidin and naringin are beneficial for improving hyperlipidemia and hyperglycemia in type-2 diabetic animals (Jung et al., 2006).
Hesperidin and naringin effectively lowered the plasma free fatty acid and plasma and hepatic triglyceride levels, and simultaneously reduced the hepatic fatty acid oxidation and carnitine palmitoyl transferase activity. Table Contd.
362
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Contd.
ActionlConclusion
AimJMethod
Naringin has a lipidlowering effect in ISOinduced MI rats (Rajadurai & Prince, 2006).
The preventive effect of naringin in isoproterenol (ISO)-induced myocardial infarction (MI) in rats was studied.
Naringin possess antilipoperoxidative and antioxidant activity in experimentally induced cardiac toxicity (Rajadurai & Prince, 2006).
The cardioprotective potential of naringin on lipid peroxides, enzymatic and nonenzymatic antioxidants and histopathological findings in isoproterenol (ISO)-induced myocardial infarction (MI) in rats were evaluated.
N aringin possess cardioprotective effect in ISO-induced MI in rats (Rajadurai & Prince, 2007).
The preventive role of naringin on cardiac troponin T (cTnT), lactate dehydrogenase (LDH)-isoenzyme, cardiac marker enzymes, electrocardiographic (ECG)-patterns and lysosomal enzymes in isoproterenol (ISO)-induced myocardial infarction (Mil in male Wistar rats were investigated. Naringin was investigated for its differential effects on hepatic cholesterol regulation when supplemented for 3 weeks and 6 weeks in Sprague-Dawley rats.
N aringin time-dependently lowers hepatic cholesterol biosynthesis and plasma cholesterol in rats fed high-fat and high-cholesterol diet (Kim et al .• 2006). N aringin has cardioprotective role in ISO-induced MI in rats (Rajadurai & Prince, 2007).
The preventive role of naringin on heart weight, blood glucose, total proteins, albumin/globulin (A/G) ratio, serum uric acid, serum iron, plasma iron binding capacity and membrane bound enzymes and glycoproteins such as hexose, hexosamine, fucose and sialic acid in isoproterenol OSO)-induced myocardial infarction (Mil in rats and in vitro free radical scavenging assay were studied.
Results Pretreatment with naringin significantly decreased the levels of total, ester, and free cholesterol, triglycerides, and free fatty acids in serum and heart and increased phospholipids in heart. Oral administration ofnaringin to ISO-induced rats showed a significant decrease in the levels of lipid peroxidative products and improved the antioxidant status. Histopathological findings of the myocardial tissue showed the protective role ofnaringin in ISO-induced rats. Pretreatment with naringin positively altered the levels of cTnT, intensity of the bands of the LDHI and LDH2-isoenzyme and the activities of cardiac marker enzymes, ECGpatterns and lysosomal hydrolases in ISO-induced rats.
Supplementation with naringin did not exhibit a hypolipidemic effect when given with a HFHC diet. Naringin can, however, be beneficial for lowering hepatic cholesterol biosynthesis and levels of plasma lipids in this animal model. Pretreatment with naringin exhibited a significant effect and altered these biochemical parameters positively in ISO-induced rats. Naringin also scavenges 1,1-diphenyl-2picrylhydrazyl,2,2'-azinobis-(3ethyl-benzothiazoline-6-sulfonic acid) and nitric oxide radicals in vitro.
Table Contd.
Naringin and its Aglycone, Research and Managements
363
Contd. Action/Conclusion
AimlMethod
(Rajadurai & Prince, The preventive role of 2007). naringin on mitochondrial enzymes in isoproterenol OSO)-induced myocardial infarction in male albino Wistar rats was studied.
Results Oral pretreatment with naringin to ISO-induced rats daily for a period of 56 days significantly minimized the alterations in all the biochemical parameters and restored the normal mitochondrial function. Transmission electron microscopic observations also correlated with these biochemical findings.
Effects on skeletal system Action/Conclusion
AimlMethod
Results
Rutin, quercetin, and naringin supplementation reduce molar crestal alveolar bone-cementoenamel junction distance in young rats (Wood,2004).
The influence of dietary bioflavonoid (rutin [R], quercetin [Ql and naringin [N]) supplementation on physiological molar crestal alveolar bone (CABl-cemento-enamel junction (CEJ) distances in young male albino rats was studied.
The N group demonstrated the lowest CAB-CEJ distance, followed by the Rand Q groups (P<.00l-.05) except in the mandibular lingual region where the Q group had a lower CABCEJ distance than the Nand R groups (P <.05). The control group showed the largest CABCEJ distances.
N aringin in collagen matrix have the effect of increasing new bone formation locally and can be used as a bone graft material (Wong & Rabie, 2006).
The amount of new bone produced by naringin in collagen matrix to that produced by bone grafts and collagen matrix in rabbits was compared.
A total of284% and 490% more new bone was present in defects grafted with naringin in collagen matrix than those grafted with bone and collagen respectively.
Besides statin, this provided another example of HMG-CoA reductase inhibition that increases the bone cell activities (Wong & Rabie, 2006).
The effect of naringin, which N aringin significantly inwas also a HMG-CoA reduc- creased bone cell activities in tase inhibitor was studied in vitro. UMR 106 osteoblastic cell line in vitro.
Two active constituents were isolated and identified as naringin and neoeriocitrin (Li et al., 2006).
The osteoblastic activity of extracts of Drynaria fortunei (Kunze) J. Sm. rhizome was assayed in the UMRl06 cell line cultured in vitro.
The ethanol extract, and its ethyl acetate and n-butanol fractions exhibited stimulating activity. Table Contd.
364
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Contd. Action/Conclusion These outcomes suggest that naringin offer a potential in the management of osteoporosis in vitro (Wei et al., 2007).
AimlMethod A retinoic acid-induced osteoporosis model of rats was used to assess whether naringin has similar bioactivity against osteoporosis in vitro.
Results A blood test showed that naringin-treated rats experienced significantly lower activity of serum alkaline phosphatase and had higher femur bone mineral density, compared to untreated rats.
Effects on smooth muscle Action/Conclusion
AimlMethod
Results
N aringin and naringlnln increased contractions induced by noradrenaline in rat vas deferens (Herrera & Marhuenda, 1993).
Effect of naringin and naringenin on contractions induced by noradrenaline in rat vas deferens was studied.
Naringin significantly increased contractions induced by noradrenaline in rat vas deferens. Naringenin increased the contractile effect of noradrenaline and was dose dependent.
Relaxant effects of flavonoids on vascular smooth muscle of the isolated rat thoracic aorta (Ajay et al., 2003).
The potency, structure-activity relationship and mechanism of vasorelaxation of flavonols: fisetin, rutin, quercetin; flavones: chrysin, flavone, baicalein; flavanones: naringenin, naringin; isoflavones: diad zein and flavanes: epigallo catechin gallate were examined in the isolated rat aorta. The mechanical and electrophysiological effects of (+/-)naringenin were investigated in vascular smooth muscle cells.
Most of the flavonoids tested showed concentration dependent relaxant effects against K+(80 mm) and phenylephrine (PE, 0.1 microMHnduced contractions with a greater inhibition of the responses to the alpha1-adrenoceptor agonist.
The vasorelaxant effect of the naturally-occurring flavonoid (+/-)naringenin on endothelium-denuded vessels was due to the activation of BK(Ca) channels in myocytes (Saponara et al.,2006).
(+/-)-Naringenin induced concentration-dependent relaxation in endothelium-denuded rat aortic rings.
Naringin and its Aglycone, Research and Managements
365
Effects on gastric intestinal system Action/Conclusion N aringenin, a specific histidine decarboxylase inhibitor, has gastric anti-ulcer activity (Parmar, 1983).
has a N aringin 'cytoprotective' effect against ethanol injury in the rat, but this property appears to be mediated by non-prostaglandin-depend en t mechanisms (Martin et al., 1994). Flavonoids promote cell migration in nontumorigenic colon epithelial cells differing in Apc genotype (Fenton & Hord, 2004).
AimlMethod
Results
The gastric anti-ulcer activity of a specific histidine decarboxylase inhibitor naringenin, has been studied on the various types of ulcers experimentally induced in rats, viz., pylorus-ligated (Shay method) and restraint ulcers, and on the gastric mucosal damage induced by aspirin, phenylbutazone or reserpine. To determine the gastroprotective properties of naringin on and the involvement of endogenous prostaglandins in mucosal injury produced by absolute ethanol.
Naringenin possessed significant anti-ulcer activity in all these models, manifesting a dose-dependent anti-ulcer effect.
Whether specific flavonoids induce cell migration in colon epithelial cells either wild type or heterozygous for Apc genotype was studied.
Naringin and hesperidin induced the greatest migratory response in IMCE cells at 1 microM and induced migration greater than untreated control cells.
Oral pretreatment with the highest dose of naringin (400 mg/kg), 60 min before absolute ethanol was the most effective antiulcer treatment.
Effects on endocrine system Action/Conclusion
AimlMethod
Dietary flavonoids in- A structure-activity study of hibit thyroid peroxidase 13 commonly consumed fla(Divi & Doerge, 1996). vonoids was conducted to evaluate inhibition of thyroid peroxidase (TPO), the enzyme that catalyzes thyroid hormone biosynthesis.
Results Inhibition by the more potent fisetin, kaempferol, naringenin, and quercetin, was consistent with mechanismbased inactivation of TPO as previously observed for resorcinol and derivatives. Myricetin and naringin inhibited TPO by different mechanisms. Table Contd.
366
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Contd. Action/Conclusion
AimlMethod
Results
N aringinin interacts with human sex hormone-binding globulin (Dechaud et al., 1999).
This study reports on some environmental chemicals with estrogenic activity (xenoestrogens) and their binding interaction for human plasma sex-hormone binding globulin (hSHBG).
The flavonoid phytoestrogens genistein and naringenin were also identified as hSHBG ligands, whereas their glucoside derivatives, genistin and naringin, had no binding activity for hSHBG.
Plants containing flavonoids may have preventive effects in diabetic complications (Asgary et aI., 2002).
Several flavonoids, such as rutin, kaempferol, quercetin, apigenin, naringin, morin and biochanin A were selected to determine their antioxidant effects on in vitro insulin, hemoglobin and albumin glycosylation.
Biochanin A, the best inhibitor of insulin and hemoglobin glycosylation, inhibits their glycosylation 100% and 60%, respectively. Glycosylation of albumin was inhibited 100% by both biochanin A and apigenin.
Hesperidin and naringin prevent the progression ofhyperglycemia, by increasing hepatic glycolysis and glycogen concentration and/or by lowering hepatic gluconeogenesis (Jung et al., 2004).
The effect of citrus bioflavonoids on blood glucose level, hepatic glucose-regulating enzymes activities, hepatic glycogen concentration, and plasma insulin levels was studied, and assessed the relations between plasma leptin and body weight, blood glucose, and plasma insulin. The effect of various doses of naringin was studied on streptozotocin (STZ)-induced hyperglycaemic rats to evaluate the possible hypoglycaemic and antioxidant activity of naringin in diabetes.
Naringin provided a significant amelioration of hypoglycaemic and antioxidant activity in STZinduced diabetic rats (Ali & Kader, 2004).
Inhibition of intestinal glucose uptake and renal glucose reabsorption explains, the antihyperglycemic action of naringenin and its derivatives (Li et al., 2006).
Hesperidin and naringin supplementation significantly reduced blood glucose compared with the control group. Naringin also markedly lowered the activity of hepatic glucose-6-phosphatase and phosphoenolpyruvate carboxykinase compared with the control group. Exogenous administration of naringin to hyperglycaemic rats causes a dose-dependent decrease of the glucose level, an increase of the insulin concentration, a decrease of the H 2 0 2 and TBARS levels, as well as the increase of the total antioxidant status. Using purified intestinal Naringenin, but not naringin, brush border membrane significantly inhibited glucose vesicles and everted intestinal uptake in the intestine. sleeves, glucose uptake in intestine was studied with naringinin and naringin.
Naringin and its Aglycone, Research and Managements
367
Effects against tumour Action/Conclusion Citrus flavonoids are effective inhibitors of human breast cancer cell proliferation in vitro, especially when paired with quercetin (So et al., 1996).
The antimutagenic properties the Citrus flavonoids, especially tangeretin and nobiletin, might prevent cancer (Calomme et al., 1996).
Foods rich in certain flavonoids may protect against certain forms of lung cancer. Decreased bioactivation of carcinogens by inhibition of CYP1A1 should be explored (Le Marchand et al.,2000). Bioflavonoids as antiradicals, antioxidants and DNA cleavage protectors(Russo et al., 2000).
AimlMethod
Results
Two citrus flavonoids, hesperetin and naringenin, and four noncitrus flavonoids, baicalein, galangin, genistein, and quercetin, were tested singly and in oneto-one combinations for their effects on proliferation and growth of a human breast carcinoma cell line, MDAMB-435. These compounds, were tested for their ability to inhibit development of mammary tumors in female Sprague-Dawley rats. The antimutagenicity of the Citrus flavonoids naringin, hesperidin, nobiletin, and tangere tin against the mutagens benzo[a]pyrene, 2aminofluorene, quercetin, and nitroquinoline N-oxide was investigated in the Salmonella/microsome assay. To investigate the possible relationship between intake of flavonoids-powerful dietary antioxidants that may also inhibit P450 enzymesand lung cancer risk, we conducted a population-based, case-control study in Hawaii.
IC 50 values for the one-to-one combinations ranged from 4.7 micrograms/mL (quercetin + hesperetin, quercetin + naringenin) to 22.5 micrograms/mL (naringenin + hesperetin). Rats given orange juice had a smaller tumor burden than controls, although they grew better than any of the other groups.
Authors investigated the free-radical scavenging capacity ofbioflavonoids (rutin, catechin, and naringin) and the effects of these polyphenols on xanthine oxidase activity, spontaneous lipid peroxidation, and DNA cleavage.
The bioflavonoids under examination showed a dose-dependent free-radical scavenging effect, a significant inhibition of xanthine oxidase activity, and an antilipoperoxidative capacity. In addition, they showed a protective effect on DNA cleavage.
N aringin and hesperidin showed a weak antimutagenic activity against benzo [a] pyrene.
Authors found statistically significant inverse associations between lung cancer risk and the main food sources of the flavonoids quercetin (onions and apples) and naringin (white grapefruit).
Table Contd.
368
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Contd. Action/Conclusion
AimlMethod
Inhibitory effects of naringenin on tumor growth in human cancer cell lines and sarcoma S-180-implanted mice (Kanno et al., 2005).
The effect ofnaringenin on tumor growth in various human cancer cell lines and sarcoma S-180-implanted mice was studied.
The tested flavonoids were weak apoptosis inducers on multidrugresistant (MDR) and parent cells (Ugocsai et al., 2005).
The effects of various flavonoids and carotenoids on Rhodamine 123 accumulation in MDR Colo 320 human colon cancer cells expressing MDRlILRP were studied. The Colo 205 cell line was used as a drug-sensitive control. The hypothesis that untreated and irradiated grapefruit as well as the isolated citrus compounds naringin and limonin would protect against azoxymethane (AOM)-induced aberrant crypt foci (ACF) by suppressing proliferation and elevating apoptosis through anti-inflammatory activities was examined.
Consumption of grapefruit or limonin may help to suppress colon cancer development (Vanamala et al., 2006).
Glycosylated flavonoids (i.e. naringin, a constituent of citrus fruits, and rutin, a constituent of cranberries) induced the greatest response to treatment at the lowest concentration in MDA human breast cancer cells (Schindler & Mentlein, 2006).
Whether secondary plant constituents, i.e. flavonoids, tocopherols, curcumin, and other substances regulate VEGF in human tumor cells in vitro was studied by measuring VEGF release by ELISA from MDA human breast cancer cells and, for comparison, U-343 and U118 glioma cells.
Results N aringenin inhibited tumor growth in sarcoma S-180-implanted mice, following intraperitoneal or peroral injection once a day for 5 d. N aringin also inhibited tumor growth by peroral injection but not intraperitoneal injection. Catechin, Neohesperidin, N aringin, Robinin, Phloridzin, Dihydrobinetin and Sakuranetin, had only marginal effects on Rhodamine 123 accumulation.
Lower levels of iNOS and COX2 are associated with suppression of proliferation and upregulation of apoptosis, which may have contributed to a decrease in the number of high multiplicity ACF in rats provided with untreated grapefruit and limonin.
The rank order ofVEGF inhibitory potency was naringin > rutin> alpha-tocopheryl succinate > lovastatin > apigenin > genistein> alpha-tocopherol >or= kaempferol > gamma-tocopherol; chrysin and curcumin were inactive except at a concentration of 100 micromollL. Glioma cells were similarly sensitive, with U343 more than U118, especially for alpha-TOS and tocopherols.
Naringin and its Aglycone, Research and Managements
369
Protections against toxins in chemotherapy drugs and the environment Action/Conclusion N aringin and other okadaic acid-antagonistic flavonoids could have potential therapeutic value as protect ants against pathological hyperphosphorylations, environmental toxins, or side effects of chemotherapeutic drugs (Gordon et ai., 1995). Action of naringin is mediated through suppression of lipopolysaccharide-induced TNF production (Kawaguchi et ai., 1999). Prevention of toxin-induced cytoskeletal disruption and apoptotic liver cell death by the grapefruit flavonoid, naringin (Blankson et ai., 2000).
Naringenin inhibits the activity of cytochrome P450 (CYP) isoforms that activate NNK (Bear & Teel, 2000a).
AimlMethod
Results
55 different flavonoids were tested for their effect on okadaic acid-inhibited autophagy, measured as the sequestration of electroinjected [3H] raffinose.
N aringin (naringenin 7hesperidoside) and several other flavanone and flavone glycosides (prunin, neoeriocitrin, neohesperidin, apiin, rhoifolin, kaempferol 3rutinosidel offered virtually complete protection against the autophagy-inhibitory effect of okadaic acid.
Suppressive effects of naringin on lipopolysaccharide-induced tumor necrosis factor (TNF) release followed by liver injury were investigated. The protein phosphatase-inhibitory algal toxins, okadaic acid and microcystin-LR, induced overphosphorylation of keratin and disruption of the keratin cytoskeleton in freshly isolated rat hepatocytes. In hepatocyte cultures, the toxins elicited DNA fragmentation and apoptotic cell death within 24 h. Authors investigated the effects of five citrus phytochemicals on the in vitro metabolism of the tobacco-specific nitrosamine NNK and on the de alkylation of methoxyresorufin (MROD) and pentoxyresorufin (PROD) in liver and lung microsomes of the Syrian golden hamster.
Treatment with naringin 3 h prior to lipopolysaccharide challenge resulted in complete protection from lipopolysaccharide lethality in D-galactosamine-sensitized mice. All these toxin effects could be prevented by the grapefruit flavonoid, naringin. The cytoprotective effect of naringin was apparently limited to normal hepatocytes, since the toxin-induced apoptosis of hepatoma cells, rat or human, was not prevented by the flavonoid. Results suggest that naringenin and quercetin from citrus fruits inhibit the activity of cytochrome P450 (CYP) isoforms that activate NNK and may afford protection against NNK-induced carcinogenesis.
Table Contd.
370
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Contd.
Action/Conclusion
AimlMethod
Diosmin, naringin, naringenin and rutin are chemoprotective towards CYPIA2 mediated mutagenesis of heterocyclic amines (HCA's) (Bear & Teel, 2000b).
Using Aroclor 1254 induced rat liver S9, four citrus flavonoids: diosmin, naringenin, naringin and rutin were tested for their effects on the mutagenicity of HCA's MeIQx, Glu-P-1*, IQ and PhIP in Salmonella typhimunum TA98.
N aringin protects against the radiation-induced genomic instability in the mice bone marrow (Jagetia & Reddy, 2002).
The effect of various doses of naringin was studied on the alteration in the radiation-induced micronucleated polychromatic (MPCE) and normochromatic (MNCE) erythrocytes in mouse bone marrow exposed to 2 Gy of 60Co gamma-radiation.
Naringin supplement regulate lipid and ethanol metabolism (Seo et al.,2003).
The effect of naringin supplements on the alcohol, lipid, and antioxidant metabolism in ethanol-treated rats was investigated.
N aringin from natural products is a useful drug having antioxidant and anti-apoptopic properties (Kanno et al., 2003).
The effects of naringin on H 2 0 2 -induced cytotoxicity and apoptosis in mouse leukemia P388 cells were investigated. H 2 0 2-induced cytotoxicity was
N aringin can protect mouse bone marrow cells against radiationinduced chromosomal damage (Jagetia et al., 2003).
The radioprotective action of 2 mg/kg naringin in the bone marrow of mice exposed to different doses of(60)Co gammaradiation was studied by scoring the frequency of asymmetrical chromosomal aberrations.
Results MeIQx induced mutagenesis and PhlP induced mutagenesis in S. typhimurium were significantly inhibited by all four flavonoids. Glu-P-1 induced mutagenesis was inhibited by rutin and naringenin. IQ induced mutagenesis was significantly inhibited by each flavonoid except diosmin. N aringin is able to protect mouse bone marrow cells against the radiation-induced DNA damage and decline in the cell proliferation as observed by a reduction in the micronucleus frequency and an increase in PCEINCE ratio, respectively, in the naringin-pretreated irradiated group. Naringin would appear to contribute to alleviating the adverse effect of ethanol ingestion by enhancing the ethanol and lipid metabolism as well as the hepatic antioxidant defense system. significantly attenuated by naringin or the reduced form of glutathione, a typical intracellular antioxidant. N aringin suppressed chromatin condensation and DNA damage induced by H 20 2 · N aringin at 5 !lM scavenged the 2,2-azino-bis-3-ethyl benzothiazoline-6-sulphonic acid cation radical very efficiently, where a 90% scavenging was observed.
Table Contd.
Naringin and its Aglycone, Research and Managements
371
Contd.
Action/Conclusion
Aim/Method
Results
Naringin and naringenin inhibit nitrite-induced methemoglobin formation (Kumar et al., 2003).
The effect of naringin and naringenin protect hemoglobin from nitrite-induced oxidation to methemoglobin was studied.
Naringenin was more effective than naringin, probably because of the extra phenolic group in the aglycone.
Suppression of infectioninduced endotoxin shock in mice by naringin (Kawaguchi et al., 2004).
The protective effect of the naringin was studied in an endotoxin shock model based on Salmonella infection. Intraperitoneal (i.p.) infection with 10 (8) CFU Salmonella typhimurium aroA caused lethaI shock in lipopolysaccharide (LPS) -responder but not LPS-non-responder mice.
Administration ofl mg naringin 3 h before infection resulted in protection from lethal shock, similar to LPS-non-responder mice. Also resulted not only in a significant decrease in bacterial numbers in spleens and livers, but also in a decrease in plasma LPS levels.
blocked N aringin apoptosis caused by AraC-induced oxidative stress, resulting in the inhibition of the cytotoxicity of Ara-C (Kanno et al.,2004).
The effect of naringin on the cytotoxicity and apoptosis in mouse leukemia P388 cells treated with Ara-C. Ara-C caused cytotoxicity in a concentration and time-dependent manner in the cells was examined. The effect of naringin, a bioflavonoid with anti-oxidant potential, was studied on Fe-NTAinduced nephrotoxicity in rats.
Naringin remarkably attenuated the Ara-C-induced apoptosis and completely blocked the DNA damage caused by Ara-C treatment at 6 h using the Comet assay.
There is a protective effect of naringin on FeNTA-induced nephrotoxicity in rats(Singh et al.,2004a).
There is a protective effect of naringin in glycerol-induced renal failure in rats (Singh et al., 2004b).
Pre-treatment of animals with naringin, 60 min before FeNTA administration, markedly attenuated renal dysfunction, morphological alterations, reduced elevated TBARS, and restored the depleted renal antioxidant enzymes. The effect ofnaringin, a biofla- Pretreatment of animals with vonoid with anti-oxidant poten- naringin 60 min prior to glyctial, was studied in glycerol-in- erol injection markedly attenuated renal dysfunction, morphoduced ARF in rats. logical alterations, reduced elevated thiobarbituric acid reacting substances (TBARS), and restored the depleted renal antioxidant enzymes. Table Contd.
372
Camp. Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
Contd.
Action/Conclusion
AimlMethod
Results
Enhanced antioxidant status by naringin could compensate the oxidative stress and may facilitate an early recovery from iron-induced genomic insult in vitro (Jagetia et al., 2004).
Whether naringin treatment may help to overcome the iron-induced toxic effects in vitro was studied.
Pretreatment of HepG2 cells with naringin resulted in an elevation in all the antioxidant enzymes.
N aringin protects mouse liver and intestine against the radiation-induced damage byelevating the antioxidant status and reducing the lipid peroxidation (Jagetia & Reddy, 2005).
The alteration in the antioxidant status and lipid peroxidation was investigated in Swiss albino mice treated with 2 mg/kg b.wt. naringin, a citrus flavoglycoside, before exposure to 0.5, 1,2, 3, and 4 Gy gamma radiation.
The alteration in the antioxidant status and lipid peroxidation was investigated in Swiss albino mice treated with 2 mg/kg b.wt. naringin, a citrus flavoglycoside, before exposure to 0.5, 1, 2, 3, and 4 Gy gamma radiation.
A combination of betacarotene with naringin, rutin or quercetin may increase the safety of beta -carotene (Yeh et al., 2005).
The interaction of beta-carotene with three flavonoidsnaringin, rutin and quercetinon DNA damage induced by ultraviolet A (UVA) in C3H10T1I2 cells was studied
All three flavonoids had some absorption at the UVA range, but the effects were opposite to those on DNA damage and beta-carotene oxidation.
Possible mechanism for the anti ocular inflammatory effect may be the suppression ofPGE 2 and NO by naringin and naringenin (Shiratori et al., 2005). Suppression of the LPSinduced mortality and production of NO by NG is due to inhibition ofthe activation ofNF-kappa~ (Kanno et al., 2006).
The efficacy of naringin and naringenin on endotoxin- induced uveitis (EIU) in rats was studied. EIU was induced in male Lewis rats by a footpad injection oflipopolysaccharide (LPS). The effect of naringin, on LPS-induced endotoxin shock in mice and NO production in RAW 264.7 macrophages was studied.
40 microMlkg of naringin and naringenin suppressed increases in cell count owing to LPS treatment by 31 % and 38%, respectively.
Naringin reduced the genotoxic effects of bleomycin and consequently increased the cell survival and therefore may act as a chemoprotective agent in clinical situations (Jagetia et al., 2005).
The effect of naringin, a grapefruit flavonone was studied on bleomycin-induced genomic damage and alteration in the survival of cultured V79 cells.
Naringin suppressed LPS -induced production of NO and the expression of inflammatory gene products such as iNOS, TNF-alpha, COX-2 and IL-6 as determined by RT-PCR assay. Treatment of cells with naringin before exposure to different concentrations of bleomycin arrested the bleomycin-induced decline in the cell survival accompanied by a significant reduction in the frequency of micronuclei when compared with bleomycin treatment alone. Table Contd.
Naringin and its Aglycone, Research and Managements
373
Contd. Action/Conclusion Among the plant ingredients tested in the current study, morin, naringin, and quercetin were found to be desirable protectors against B[a]P photo toxicity(Rori et al., 2007).
AimIMethod A diversity of antioxidants and plant ingredients were examined for their protective effect in cultured Balb/c 3T3 cells against ultraviolet A (UVA)-induced cytotoxicities of extracted air pollutants and benz[a]pyrene (B[a]P).
Results The B[a]P phototoxicity was not eliminated by well-known antioxidants but was markedly diminished by diversity of plant ingredients.
Antioxidant effects Action/Conclusion Some flavonoids have inhibitory action on enhanced spontaneous lipid peroxidation following glutathione depletion (Younes & Siegers, 1981).
Citrus fruit peels have antioxidant activity (Kroyer, 1986).
Flavonoids have protective effect against lipid peroxidation of erythrocyte membranes (Affany et al., 1987).
AimlMethod Depletion of hepatic glutathione in phenobarbitalinduced rats by phorone (diisopropylidene acetone) led to an enhancement of spontaneous lipid peroxidation in vitro. Addition of exogenous glutathione, dithiocarb or one of the flavonoids (+ )-catechin, (- )-epica techin, 3-0methylcatechin, quercetin, taxifolin, rutin, naringin or naringenin led in every case to a dose-dependent inhibition of this peroxidative activity. The antioxidant properties of freeze-dried citrus fruit peels (orange, lemon, grapefruit) and methanolic extracts from the peel were studied. Cumene hydroperoxide induces in vitro the peroxidation of erythrocyte membrane. The protective effect of various flavonoids was compared to that of butylated hydroxy toluene (BHT). Protective effect was evaluated by the inhibition of peroxidation product formation.
Results The concentration values yielding 50% inhibition (I (50)) varied from 1.0 x 10 (-6) M for glutathione to 1.9 x 10 (-5) M for naringenin.
Freeze-dried orange peel showed the highest, lemon peel somewhat less and grapefruit peel the lowest but still remarkable antioxidant activity. Quercetin and catechin showed a protective effect against lipid peroxidation as high as that of BHT. Morin, rutin, trihydroxyethylrutin, and naringin were active but to a lesser degree.
374
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Contd. Action/Conclusion Polyhydroxylated substitutions on rings A and B, a 2,3-double bond, a free 3-hydroxyl substitution and a 4keto moiety confer antiperoxidative properties (Ratty & Das, 1988). Flavonoids are superoxide scavengers and antioxidants (Chen et al., 1990).
Aromatic hydroxyl group is very important for antioxidative effects of the compounds. None of the compounds tested exerted an obvious prooxidant effect (Ng et al., 2000).
Antioxidative activity of naringin and lovastatin in high cholesterol-fed rabbits (Jeon et al., 200n
AimlMethod
Results
The in vitro effects of several flavonoids on nonenzymatic lipid peroxidation in the rat brain mitochondria was studied. The lipid peroxidation was indexed by using the 2thiobarbituric acid test.
The flavonoids, apigenin, flavone, flavanone, hesperidin, naringin, and tangeretin promoted the ascorbic acid-induced lipid peroxidation.
The superoxide anions scavenging activity and antioxidation of seven flavonoids were studied. The superoxide anions were generated in a phenazin methosulphate-NADH system and were assayed by reduction of nitroblue tetrazolium. A variety of flavonoids, lignans, an alkaloid, a bisbenzyl, coumarins and terpenes isolated from Chinese herbs was tested for antioxidant activity as reflected in the ability to inhibit lipid peroxidation in rat brain and kidney homogenates and rat erythrocyte hemolysis. The pro-oxidant activities of the aforementioned compounds were assessed by their effects on bleomycin-induced DNA damage. To determine the antioxidative effects of the citrus bioflavonoid, naringin, a potent cholesterol-lowering agent, compared to the cholesterol-lowering drug, lovastatin, in rabbits fed a high cholesterol diet.
The scavenging activity ranked: rutin was the strongest, and quercetin and naringin the second, while morin and hispidulin were very weak.
The flavonoid rutin and the terpene tanshinone I manifested potent antioxidative activity in the lipid peroxidation assay but no inhibitory activity in the hemolysis assay. The lignan deoxypodophyllotoxin, the flavonoid naringin and the coumarins columbianetin, bergapten and angelicin slightly inhibited lipid peroxidation in brain and kidney homogenates.
N aringin regulate antioxidative capacities by increasing the SOD and catalase activities, up-regulating the gene expressions of SOD, catalase, and GSH-Px, and protecting the plasma vitamin E. Lovastatin exhibited an inhibitory effect on the plasma and hepatic lipid peroxidation and increased the hepatic catalase activity. Table Contd.
Naringin and its Aglycone, Research and Managements
375
Contd.
Action/Conclusion
AimlMethod
Results
Antioxidant effects of naringin and probucol in cholesterol-fed rabbits (Jeon et al., 2002).
Twenty male rabbits were served a high-cholesterol diet or high-cholesterol diet supplemented with naringin or probucol for 8 weeks to compare the antioxidative effects of the naringin and antioxidative cholesterollowering drug (probucol).
The probucol supplement was very potent in the antioxidative defense system, whereas naringin exhibited a comparable antioxidant capacity based on increasing the gene expressions in the antioxidant enzymes, increasing the hepatic SOD and CAT activities, sparing plasma vitamin E and decreasing the hepatic mitochondrial H 20 2 content.
Several structural features were linked to the strong antioxidant activity of flavonoids (Yu et al., 2005).
A variety of in vitro models such as beta-carotene-linoleic acid, 1,1-diphenyl-2picryl hydrazyl (DPPH), superoxide, and hamster lowdensity lipoprotein (LDL) were used to measure the antioxidant activity of 11 citrus bioactive compounds. The influence of naringin versus red grapefruit juice on plasma lipid levels and plasma antioxidant activity in rats fed cholesterol-containing and cholesterol-free diets was compared.
Flavonoids, which contain a chromanol ring system had stronger antioxidant activity as compared to limonoids and bergapten, which lack the hydroxy groups.
N aringin is a powerful plasma lipid lowering and plasma antioxidant activity increasing flavonone. However, fresh red grapefruit is preferable than naringin (Gorinstein et al., 2005). Induction of cell apoptosis in 3T3-L1 preadipocytes by flavonoids is associated with their antioxidant activity(Hsu & Yen 2006). The Naringin-Cu (II) complex 1 showed higher antioxidant, anti-inflammatory and tumor cell cytotoxicity activities than free naringin without reducing cell viability(Pereira et al., 2007).
The relationship between the influence offlavonoids on cell population growth and their antioxidant activity was studied. In order to understand the contribution of the metal coordination and the type of interaction between a flavonoid and the metal ion, in this study a new metal complex of Cu (II) with naringin was synthesized and characterized by FT-IR, UV-VIS, mass spectrometry (ESI-MS/ MS), elemental analysis and 1H-NMR.
After 30 days of different feeding, it was found that diets supplemented with red grapefruit juice and to a lesser degree with naringin improved the plasma lipid levels mainly in rats fed cholesterol and increased the plasma antioxidant activity. The inhibition of flavonoids (naringenin, rutin, hesperidin, resveratrol, naringin and quercetin) on 3T3-L1 pre-adipocytes was 28.3, 8.1, 11.1, 33.2, 5.6 and 71.5%, respectively. The results of these analyses indicate that the complex has a Cu (II) ion coordinated via positions 4 and 5 of the flavonoid.
376
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Antimicrobial effects ActionlConclusion
Method
Results
Naringin inhibited P. Coumarins, flavonoids and aeruginosa (Ng et al., polysaccharopeptide were tested for antibacterial activity. 1996). The bacteria used for this study included clinical isolates of Staphylococcus aureus, Shigella flexneri, Salmonella typhi, Escherichia coli and Pseudomonas aeruginosa.
When tested at the dose of 128 mg/l, the flavonoids (rutin, naringin and baicalin) inhibited 25% or less of P. aeruginosa and only baicalin was active against S. aureus.
Anti-Sindbis activity of flavanones hesperetin and naringenin (Paredes et al., 2003).
The effect of hesperetin, naringenin and its glycoside form on the Sindbis neurovirulent strain (NSV) replication in vitro was studied. All flavanones tested were not cytotoxic on Baby Hamster cells 21 clone 15 (BHK-21l.
Hesperetin and naringenin had inhibitory activity on NSV infection. However their glycosides, hesperidin and naringin did not have inhibitory activity. Implying that the presence of rutinose moiety offlavanones blocks the antiviral effect.
There exist antimicrobial activity of grapefruit seed and pulp ethanolic extract(Cvetnic & Vladimir-Knezevi c, 2004).
Antibacterial and antifungal activity of ethanolic extract of grapefruit (Citrus paradisi Macf., Rutaceae) seed and pulp was examined against 20 bacterial and 10 yeast strains.
Ethanolic extract exibited the strongest antimicrobial effect against Salmonella enteritidis (MIC 2.06%, roN). Other tested bacteria and yeasts were sensitive to extract concentrations ranging from 4.13% to 16.50%
Selected bioflavonoids may show promise as an alternative means of reducing dental caries (Wood,2007).
The present study evaluates two separate, but related, dietary trials-trial 1, dietary naringenin (NAR) supplementation; and trial 2, dietary rutin (R), quercetin (Q), and naringin (N) supplementationon dental caries formation in 40 different male albino rats, at the expense of dextrose, for periods of 42 days.
An inverse dose-dependent relationship was established among the NAR experimental groups and control group. In dietary trial 2, statistically significant reductions in occlusal caries were observed for R, Q, and N in the maxillary molars and for Q and N in the mandibular molars compared with the control group.
Naringin possesses significant antimicrobial properties on periodontal pathogens in vitro. It also has an inhibitory effect on some common oral microorganisms in low concentrations (Tsui et al., 2007).
The effects of naringin on the N aringin also had an inhibitory growth of periodontal patho- effect against all bacteria and gens such as A. yeasts tested. actinomycetemcomitans and P. gingivalis were studied in vitro. For comparison, the effects of naringin on several oral microbes were also studied.
(roN).
Naringin and its Aglycone, Research and Managements
377
Drug interactions Action/Conclusion
Method
Grapefruit juice and naringenin inhibit CYP1A2 activity in man (Fuhr et al., 1993).
The effects of grapefruit juice and naringenin on the activity of the human cytochrome P450 isoform CYP1A2 were evaluated using caffeine as a probe substrate.
Grapefruit juice produces a marked and variable increase in felodipine bioavailability (Bailey et al., 1993a).
The pharmacokinetics of felodipine and its single primary oxidative metabolite, dehydrofelodipine, were studied after drug administration with 200 mL water, grapefruit juice, or naringin in water at the same concentration as the juice in a randomized crossover trial of nine healthy men. The pharmacokinetics of nisoldipine coat-core tablet were studied in a Latin squaredesigned trial in which 12 healthy men were administered the drug with water, grapefruit juice, or encapsulated naringin powder at the same amount as that assayed in the juice. To investigate whether the presence of naringin is demanded for the inhibition of the coumarin 7-hydroxylase in man or other compounds are responsible for it.
The bioavailability of some dihydropyridine calcium antagonists can be augmented by grapefruit juice but does not involve naringin (Bailey et al., 1993b).
As naringin alone is in-
effective, the inhibitory effect of grapefruit juice on the metabolism of coumarin is caused by at least one compound other than naringin (Runkel et al., 1997). To avoid the interaction, nimodipine should not be taken with grapefruit juice (Fuhr et al., 1998).
Results In vitro naringin was a potent competitive inhibitor of caffeine 3-demethylation by human liver microsomes (Ki 7-29 microM). In vivo grapefruit juice decreased the oral clearance of caffeine and prolonged its half-life. Grapefruit juice produces a marked and variable increase in felodipine bioavailability.Naringin solution produced much less of an interaction, showing that other factors were important.
The bioavailability of some dihydropyridine calcium antagonists can be markedly augmented by grapefruit juice. The naringin capsule did not change nisoldipine pharmacokinetics.
While increasing amounts of grapefruit juice delay the excretion of 7-hydroxycoumarin by 2 h, increasing doses of naringin in water up to twofold do not cause any alteration in the time course of excretion.
A randomized crossover inter- Grapefruit juice increases oral action study on the effects of nimodipine bioavailability. grapefruit juice on the pharmacokinetics of nimodipine and its metabolites. Table Contd.
378
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Contd. Action/Conclusion
Method
Results
Naringin and 6',7'dihydroxybergamottin are not the major active ingredients, although they may contribute to the grapefruit juicefelodipine interaction (Bailey et ai., 1998).
To test whether naringin or 6',7'-dihydroxybergamottin is a major active substance in grapefruit juice-felodipine interaction in humans.
The findings show the importance of in vivo testing to determine the ingredients in grapefruit juice responsible for inhibition of cytochrome P450 3A4 in humans.
In vitro inhibition of simvastatin (SV) metabolism in rat and human liver by naringenin (NRG) (Ubeaud et al., 1999). These effects may indicate a chemopreventive role of naringin against protoxicants activated by P450 1A2 (Ueng et al., 1999). Patients taking drugs which are P-glycoprotein substrates may need to restrict their intake of bioflavonoidcontaining foods and beverages, such as grapefruit juice (Mitsunaga et al., 2000).
NRG's inhibition of the metabolism of SV in rat hepatocytes (the intrinsic clearance of SV) was studied.
Naringenin present in grapefruit juice inhibits in vitro the metabolism of simvastatin, a HMG-CoA reductase inhibitor.
In vitro and in vivo effects of naringin on microsomal monooxygenase were studied to evaluate the drug interaction of this flavonoid.
Naringenin is a potent inhibitor ofbenzo(a)pyrene hydroxylase activity in vitro and naringin reduces the P450 1A2 protein level in vivo.
To see whether grapefruit juice bioflavonoids alter the permeation of vincristine across the blood-brain barrier, we conducted experiments with cultured mouse brain capillary endothelial cells (MBEC4 cells) in vitro and ddY mice in vivo.
The in vivo brain-to-plasma concentration ratio of [3Hlvincristine in ddY mice was decreased by coadministration of 0.1 mg/kg quercetin, but increased by 1.0 mg/kg quercetin. Kaempferol had a similar biphasic effect. Cchrysin, flavon, hesperetin, naringenin increased [3Hlvincristine uptake in the 10-50 microM range and glycosides (hesperidin, naringin, rutin) were without effect. Quercetin, genistein, naringin, diosmin, acacetin and chrysin increased uptake of [14Cl cefixime dose dependently by up to 60%.
Flavonoids with EGFreceptor tyrosine kinase inhibitory activities enhance the intestinal absorption of the betalactam antibiotic cefixime in Caco-2 cells.
33 flavonoids, occurring ubiquitously in foods of plant origin were tested for their ability to alter the transport of the beta-Iactam antibiotic cefixime via the H+-coupled intestinal peptide transporter PEPT1 in the human intestinal epithelial cell line Caco-2.
Table Contd.
Naringin and its Aglycone, Research and Managements
379
Contd. ActionlConclusion
Method
Results
Bergapten appears to be a potent inhibitor of CYP3A4 and may therefore be primarily responsible for the effect of grapefruit juice on CYP3A4 activity (Ho et at., 2001).
To evaluate the inhibition of CYP3A4 activity in human liver microsomes by flavonoids, furanocoumarins and related compounds and investigate possibly more important and potential inhibitors of CYF3A4 in grapefruit juice.
Bergapten (5methoxypsoralen) with the lowest IC 50 value (19-36 microM) was the most potent CYF3A4 inhibitor.
Enhanced paclitaxel bioavailability after oral coadministration of paclitaxel prodrug with naringin to rats (Choi & Shin, 2005).
The effect of naringin on the bioavailability and pharmacokinetics of paclitaxel after oral administration ofpaclitaxel or its prodrug coadministered with naringin to rats was studied. The pharmacokinetics of verapamil and one of its metabolites, norverapamil were investigated after oral administration ofverapamil at a dose of9 mg/kg without or with oral naringin at a dose of 7.5 mg! kg in rabbits.
The bioavailability ofpaclitaxel coadministered as a prodrug with or without naringin was remarkably higher than the control.
The metabolism of verapamil and the formation of norverapamil were inhibited by naringin possibly by inhibition of CYP3A in rabbits (Kim & Choi, 2005). The concomitant use of naringin significantly enhanced the oral exposure of diltiazem in rats (Choi & Han, 2005).
Verapamil dosage should be adjusted when given with naringin or a naringincontaining dietary supplement (Yeum & Choi, 2006). The inhibition of hepatic P-gp by oral naringin could also contribute to the significantly greater AUC of intravenous paclitaxel by oral naringin (Lim & Choi, 2006).
With naringin, the total area under the plasma concentration-time curve (AUC) of verapamil was significantly greater, the AUC(verapamil)1 AUC(norverapamil) ratio was considerably greater.
Pharmacokinetic parameters of diltiazem and desacetyldiltiazem were determined in rats following an oral administration of diltiazem to rats in the presence and absence of naringin. The effect of naringin on the pharmacokinetics of verapamil and its major metabolite, norverapamil in rabbits were studied.
Absolute and relative bioavailability values of diltiazem in the presence of naringin were significantly higher than those from the control group.
The effects of oral naringin on the pharmacokinetics of intravenous paclitaxel in rats were studied.
After intravenous administration of paclitaxel, the AUC was significantly greater and CI was significantly slower than controls.
Pretreatment of naringin enhanced the oral bioavailability ofverapamil.
Table Contd.
380
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Contd. Action/Conclusion
Method
Results
Naringin is a major and selective clinical inhibitor of organic aniontransporting polypeptide lA2 (OATPIA2) in grapefruit juice (Bailey et al., 2007).
Inhibition of OATPIA2 transport by flavonoids in grapefruit (naringin) and orange (hesperidin) was conducted in vitro. Two randomized crossover pharmacokinetic studies were performed clinically.
Naringin most probably directly inhibited enteric OATPIA2 to decrease oral fexofenadine bioavailability. InactivationofentericCYF3A4 was probably not involved.
Kaempferol and naringenin are shown to mediate pharmacokinetic drug interaction with the prodrugs lovastatin and enalapril due to their capability of esterase inhibition (Li et al., 2007).
The esterase-inhibitory potential of 10 constitutive flavonoids and furanocoumarins toward p-nitrophenylacetate (PNPA) hydrolysis was investigated.
In Caco-2 cells, demonstrated to contain minimal CYF3A activity, the permeability coefficient ofthe prodrugs lovastatin and enalapril was increased in the presence of the active flavonoids kaempferol and naringenin, consistent with inhibition of esterase activity.
The in vitro data suggest that compounds present in grapefruit juice are able to inhibit the P-gp activity modifYing the disposition of drugs that are P-gp substrates such as talinolol (de Castro et al., 2007).
The potential interaction between selected ingredients of grapefruit juice and, the transport of talinolol, a P-gp substrate, across Caco-2 cells monolayers was determined in the absence and presence of distinct concentrations of grapefruit juice, bergamottin, 6',7' -dihydroxybergamottin, 6',7'-epoxybergamottin, naringin and naringenin. The cellular uptake of benzoic acid was examined in the presence and the absence of naringin, naringenin, morin, silybin and quercetin in Caco-2 cells.
The flavonoid aglycone naringenin was around 10-fold more potent than its glycoside naringin with IC 50 values of236 and 2409 microM, respectively.
Some flavonoids appeared to be competitive inhibitors of mono carboxylate transporter 1 (MCTl) (Shim et al., 2007).
All the tested flavonoids except naringin significantly inhibited the cellular uptake of [(14)Clbenzoic acid. Particularly, naringenin and silybin exhibited strong inhibition effects.
Naringin and its Aglycone, Research and Managements
381
Antiinflammatory effects Action/Conclusion Flavonoids with antioxidant action (naringin and rutin) prevent the release of mastocytic and nonmastocytic histamine (Lambev et al., 1980a).
Naringin has antiexudative effect of in experimental pulmonary edema and peritonitis (Lambev et al., 1980b).
Flavonoid inhibited human basophil histamine release stimulated by various agents (Middleton & Drzewiecki, 1984).
Flavanone glycosides can be activated by intestinal bacteria, and may be effective toward IgE-induced atopic allergies (Park et al., 2005).
Method
Results
Experiments are carried out on 35 male albino rats. The effect of the flavonoids naringin and rutin on the level of mastocytic and nonmastocytic histamine is studied, as well as on its release induced by compound 48/ 80 (2 mg/kg i. p.l. The histamine content is determined fluorimetrically. The authors examined antiexudative activity of bioflavonoids naringin and rutin in comparative aspect in two models of acute inflammation. The experiments were carried out on 180 male white rats and 24 guinea pigs. Eleven flavonoids included flavone, quercetin, taxifolin, chalcone, apigenin, fisetin, rutin, phloretin tangeretin, hesperetin and naringin were studied for their effects on human basophil histamine release triggered by six different stimuli.
Naringin and rutin have no effect on the levels of mastocytic and nonmastocytic histamine. They prevent the release of mastocytic histamine, induced by compound 48/80.
The passive cutaneous anaphylaxis-inhibitory activity of the flavanones isolated from the pericarp of Citrus unshiu (Family Rutaceae) and the fruit of Poncirus trifoliata (Family Rutaceae) was studied.
Naringenin, hesperetin and ponciretin potently inhibited IgE-induced beta-hexosaminidase release from RBL-2H3 cells and the peA reaction.
The two flavonoids manifested marked antiexudative effect in rats with experiments peritonitis.
The flavonols, quercetin and fisetin, and the flavone, apigenin, exhibited a predilection to inhibit histamine release stimulated by IgE-dependent ligands (antigen, anti-IgE and con A). The flavanone derivatives, taxifolin and hesperetin, were inactive as were the glycosides, rutin and naringin. The open chain congeners chalcone and phloretin, also possessed inhibitory activity.
382
Comp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Other effects Action/Conclusion Naringin and naringenin have been identified as prooxidants independent of transition metal catalysed autoxidation reactions (Chan et al., 1999).
Method Flavonoids containing phenol B rings, e.g. naringenin, naringin, hesperetin and apigenin, were studied if they formed prooxidant metabolites that oxidised NADH upon oxidation by peroxidaseiH 2 0 2 .
Results Prooxidant phenoxyl radicals formed by these flavonoids cooxidise NADH to form NAD radicals which then activated oxygen.
REFERENCES Affany, A., Salvayre, R. and Douste-Blazy, L. (1987). Comparison of the protective effect of various flavonoids against lipid peroxidation of erythrocyte membranes (induced by cumene hydroperoxide). Fundam Clin Pharmacal., 1(6): 451-7. Ajay, M., Gilani, A.D. and Mustafa, M.R. (2003). Effects of flavonoids on vascular smooth muscle of the isolated rat thoracic aorta. Life Sci., 74(5): 603-12. Ali, M.M. and EI Kader, M.A. (2004). The influence of naringin on the oxidative state of rats with streptozotocin-induced acute hyperglycaemia. Z Naturfarsch [C], 59(9-10): 726-33. Ameer, B., Weintraub, R.A., Johnson, J.V., Yost, R.A. and Rouseff, R.L. (1996). Flavanone absorption after naringin, hesperidin, and citrus administration. Clin Pharmacol Ther., 60(1): 34-40. Asgary, S., Naderi, G.A., Zadegan, N.S. and Vakili, R. (2002). The inhibitory effects of pure flavonoids on in vitro protein glycosylation. J Herb Pharmacother., 2(2): 47-55. Bailey, D.G., Arnold, J.M., Munoz, C. and Spence, J.D. (1993a). Grapefruit juice-felodipine interaction: mechanism, predictability, and effect ofnaringin. Clin Pharmacal Ther., 53(6): 637-42. Bailey, D.G., Arnold, J.M., Strong, H.A., Munoz, C. and Spence, J.D. (1993b). Effect of grapefruit juice and naringin on nisoldipine pharmacokinetics. Clin Pharmacol Ther., 54(6): 589-94. Bailey, D.G., Dresser, G.K, Leake, B.F. and Kim, R.B. (2007). Naringin is a major and selective clinical inhibitor of organic anion-transporting polypeptide lA2 (OATPIA2) in grapefruit juice. Clin Pharmacol Ther., 81(4): 495-502. Bailey, D.G., Kreeft, J.H., Munoz, C., Freeman, D.J. and Bend, J.R. (1998). Grapefruit juicefelodipine interaction: effect of naringin and 6', 7' -dihydroxybergamottin in humans. Clin Pharmacol Ther., 64(3): 248-56. Bear, W.L. and Teel, R.W. (2000a.) Effects of citrus phytochemicals on liver and lung cytochrome P450 activity and on the in vitro metabolism ofthe tobacco-specific nitrosamine NNK. Anticancer Res., 20(5A): 3323-9. Bear, W.L. and Teel, R.W. (2000b). Effects of citrus flavonoids on the mutagenicity of heterocyclic amines and on cytochrome P450 lA2 activity. Anticancer Res., 20(5B): 360914. Blankson, H., Grotter~d, E.M. and Seglen, P.O. (2000). Prevention of toxin-induced cytoskeletal disruption and apoptotic liver cell death by the grapefruit flavonoid, naringin. Cell Death Differ., 7: 739-746. Bok, S.H., Lee, S.H., Park, Y.B., Bae, KH., Son, KH., Jeong, T.S. and Choi, M.S. (1999). Plasma and hepatic cholesterol and hepatic activities of 3-hydroxy-3-methyl-glutarylCoA reductase and acyl CoA: cholesterol transferase are lower in rats fed citrus peel extract or a mixture of citrus bioflavonoids. J Nutr., 129(6): 1182-5. Calomme, M., Pieters, L., Vlietinck, A. and Vanden Berghe, D. (1996). Inhibition of bacterial mutagenesis by citrus flavonoids. Planta Med., 62(3): 222-6.
Naringin and its Aglycone, Research and Managements
383
Chan, T., Galati, G. and O'Brien, P.J. (1999). Oxygen activation during peroxidase catalysed metabolism of flavones or flavanones. Chem Bioi Interact., 122(1): 15-25. Chen, Y.T., Zheng, R.L., Jia, Z.J. and Ju, Y. (1990). Flavonoids as superoxide scavengers and antioxidants. Free Radic Bwl Med., 9(1): 19-2l. Chiou, G.C. and Xu, X.R. (2004). Effects of some natural flavonoids on retinal function recovery after ischemic insult in the rat. J Ocul Pharmacol Ther., 20(2): 107-13. Choe, S.C., Kim, H.S., Jeong, T.S., Bok, S.H. and Park, Y.B. (2001). Naringin has an anti atherogenic effect with the inhibition of intercellular adhesion molecule-1 in hypercholesterolemic rabbits. J Cardiouasc Pharmacol., 38(6): 947-55. Choi, M.S., Do, KM., Park, Y.S., Jeon, S.M., Jeong, T.S., Lee, Y.K, Lee, M.K and Bok, S.H. (2001). Effect of naringin supplementation on cholesterol metabolism and antioxidant status in rats fed high cholesterol with different levels of vitamin E. Ann Nutr Metab., 45(5): 193-20l. Choi, J.S. and Han, H.K (2005). Enhanced oral exposure of diltiazem by the concomitant use of naringin in rats. Int J Pharm., 305(1-2): 122-8. Choi, J.S. and Shin, S.C. (2005). Enhanced paclitaxel bioavailability after oral coadministration of paclitaxel prodrug with naringin to rats. Int J Pharm., 292(1-2): 149-56. Cvetnic, Z. and Vladimir-Knezevic, S. (2004). Antimicrobial activity of grapefruit seed and pulp ethanolic extract. Acta Pharm., 54(3): 243-50. da Silva, R.R., de Oliveira, T.T., Nagem, T.J., Pinto, A.S., Albino, L.F., de Almeida, M.R., de Moraes, G.H. and Pinto, J.G. (2001). Hypocholesterolemic effect of naringin and rutin flavonoids. Arch Latinoam Nutr., 51(3): 258-64. de Castro, W.V., Mertens-Talcott, S., Derendorf, H. and Butterweck, V. (2007). Grapefruit juice-drug interactions: Grapefruit juice and its components inhibit P-glycoprotein (ABCB1) mediated transport of talinolol in Caco-2 cells. J Pharm Sci., 96(10): 2808-17. Dechaud, H., Ravard, C., Claustrat, F., de la Perriere, A.B. and Pugeat, M. (1999). Xenoestrogen interaction with human sex hormone-binding globulin (hSHBG). Steroids, 64(5): 328-34. Divi, R.L. and Doerge, D.R. (1996). Inhibition of thyroid peroxidase by dietary flavonoids. Chem Res Toxicol., 9(1): 16-23. Fenton, J.1. and Hord, N.G. (2004). Flavonoids promote cell migration in nontumorigenic colon epithelial cells differing in Apc genotype: implications of matrix metalloproteinase activity. Nutr Cancer, 48(2): 182-8. Fuhr, U., Klittich, K and Staib, A.H. (1993). Inhibitory effect of grapefruit juice and its bitter principal, naringenin, on CYP1A2 dependent metabolism of caffeine in man. Br J Clin Pharmacol., 35(4): 431-6. Fuhr, U., Maier-Briiggemann, A., Blume, H., Muck, W., Unger, S., Kuhlmann, J., Huschka, C., Zaigler, M., Rietbrock, S. and Staib, A.H. (1998). Grapefruit juice increases oral nimodipine bioavailability. Int J Clm Pharmacol Ther., 36(3): 126-32. Gordon, P.B., Holen, I. and Seglen, P.O. (1995). Protection by naringin and some other flavonoids of hepatocytic autophagy and endocytosis against inhibition by okadaic acid. J Bioi Chem., 270(11): 5830-8. Herrera, M.D. and Marhuenda, E. (1993). Effect of naringin and naringenin on contractions induced by noradrenaline in rat vas deferens-I. Evidence for postsynaptic alpha-2 adrenergic receptor. Gen Pharmacol., 24(3): 739-42. Ho, P.C., Saville, D.J. and Wanwimolruk, S. (2001). Inhibition of human CYP3A4 activity by grapefruit flavonoids, furanocoumarins and related compounds. J Pharm Pharm Sci., 4(3): 217-27. Hori, M., Kojima, H., Nakata, S., Konishi, H., Kitagawa, A. and Kawai, K (2007). A search for the plant ingredients that protect cells from air pollutants and benz[a]pyrene phototoxicity. Drug Chem Toxicol., 30(2): 105-16. Hsiu, S.L., Huang, T.Y., Hou, Y.C., Chin, D.H. and Chao, P.D. (2002). Comparison of metabolic pharmacokinetics of naringin and naringenin in rabbits. Life Sci., 70(13): 1481-9. Hsu, C.L. and Yen, G.C. (2006). Induction of cell apoptosis in 3T3-L1 pre-adipocytes by flavonoids is associated with their antioxidant activity. Mol Nutr Food Res., 50(11): 1072-9.
384
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Jagetia, A., Jagetia, G.C. and Jha, S. (2007). Naringin, a grapefruit flavanone, protects V79 cells against the bleomycin-induced genotoxicity and decline in survival. J Appl Toxicol., 27(2): 122-32. Jagetia, G.C. and Reddy, T.K (2002). The grapefruit flavanone naringin protects against the radiation-induced genomic instability in the mice bone marrow: a micronucleus study. Mutat Res., 519(1-2): 37-48. Jagetia, G.C. and Reddy, T.K (2005). Modulation of radiation-induced alteration in the antioxidant status of mice by naringin. Life SCI., 77(7): 780-94. Jagetia, G.C., Reddy, T.K, Venkatesha, V.A. and Kedlaya, R. (2004). Influence of naringin on ferric iron induced oxidative damage in vitro. Clin Chim Acta., 347(1-2): 189-97. Jagetia, G.C., Venkatesha, V.A. and Reddy, T.K (2003). Naringin, a citrus flavonone, protects against radiation-induced chromosome damage in mouse bone marrow. Mutagenesis., 18(4): 337-43. Jeon, S.M., Bok, S.H., Jang, M.K, Kim, Y.H., Nam, KT., Jeong, T.S., Park, Y.B. and Choi, M.S. (2002). Comparison of antioxidant effects of naringin and probucol in cholesterolfed rabbits. Clm Chim Acta., 317(1-2): 181-90. Jeon, S.M., Bok, S.H., Jang, M.K, Lee, M.K, Nam, KT., Park, Y.B., Rhee, S.J. and Choi, M.S. (2001). Antioxidative activity of naringin and lovastatin in high cholesterol-fed rabbits. Life Sci., 69(24): 2855-66. Jeon, S.M., Park, Y.B. and Choi, M.S. (2004). Antihypercholesterolemic property ofnaringin alters plasma and tissue lipids, cholesterol-regulating enzymes, fecal sterol and tissue morphology in rabbits. Clin Nutr., 23(5): 1025-34. Jung, U.J., Kim, H.J., Lee, J.8., Lee, M.K, Kim, H.O., Park, E.J., Kim, H.K, Jeong, T.S. and Choi, M.S. (2003). Naringin supplementation lowers plasma lipids and enhances erythrocyte antioxidant enzyme activities in hypercholesterolemic subjects. Clin Nutr., 22(6): 561-8. Jung, U.J., Lee, M.K, Park, Y.B., Kang, M.A. and Choi, M.S. (2006). Effect of citrus flavonoids on lipid metabolism and glucose-regulating enzyme mRNA levels in type-2 diabetic mice. Int J Biochem Cell Biol., 38(7): 1134-45. Jung, U.J., Lee, M.K, Jeong, KS. and Choi, M.S. (2004). The hypoglycemic effects of hesperidin and naringin are partly mediated by hepatic glucose-regulating enzymes in C57BLlKsJ-db/db mice. J Nutr., 134(10): 2499-503. Kanaze, F.l., Bounartzi, M.l., Georgarakis, M. and Niopas, 1. (2007). Pharmacokinetics of the citrus flavanone aglycones hesperetin and naringenin after single oral administration in human subjects. Eur J Clm Nutr., 61(4): 472-7. Kanno, S., Shouji, A., Asou, K and Ishikawa, M. (2003). Effects of naringin on hydrogen peroxide-induced cytotoxicity and apoptosis in P388 cells. J Pharmacol SC!., 92(2): 166-70. Kanno, S., Shouji, A., Hirata, R., Asou, K and Ishikawa, M. (2004). Effects of naringin on cytosine arabinoside (Ara-C)-induced cytotoxicity and apoptosis in P388 cells. Life Sci., 75(3): 353-65. Kanno, S., Shouji, A., Tomizawa, A., Hiura, T., Osanai, Y., Ujibe, M., Obara, Y., Nakahata, N. and Ishikawa, M. (2006). Inhibitory effect of naringin on lipopolysaccharide (LPS)induced endotoxin shock in mice and nitric oxide production in RAW 264.7 macrophages. Life Sci., 78(7): 673-8l. Kanno, S., Tomizawa, A., Hiura, T., Osanai, Y., Shouji, A., Ujibe, M., Ohtake, T., Kimura, K and Ishikawa, M. (2005). Inhibitory effects of naringenin on tumor growth in human cancer cell lines and sarcoma S-180-implanted mice. Biol Pharm Bull., 28(3): 527-30. Kawaguchi, K, Kikuchi, S., Hasegawa, H., Maruyama, H., Morita, H. and Kumazawa, Y. (1999). Suppression of lipopolysaccharide-induced tumor necrosis factor-release and liver injury in mice by naringin. Eur J Pharmacol., 368(2-3): 245-50. Kawaguchi, K, Kikuchi, S., Hasunuma, R., Maruyama, H., Ryll, R. and Kumazawa, Y. (2004). Suppression of infection-induced endotoxin shock in mice by a citrus flavanone naringin. Planta Med., 70(1): 17-22. Kim, H.J. and Choi, J.S. (2005). Effects of naringin on the pharmacokinetics of verapamil and one of its metabolites, norverapamil, in rabbits. Biopharm Drug Dispos., 26(7): 295-300.
Naringin and its Aglycone, Research and Managements
385
Kim, S.Y., Kim, H.J., Lee, M.K, Jeon, S.M., Do, G.M., Kwon, E.Y., Cho, Y.Y., Kim, D.J., Jeong, KS., Park, Y.B., Ha, T.Y. and Choi, M.S. (2006). Naringin time-dependently lowers hepatic cholesterol biosynthesis and plasma cholesterol in rats fed high-fat and highcholesterol diet. J Med Food, 9(4): 582-6. Kim, H.J., Oh, G.T., Park, Y.B., Lee, M.K., Seo, H.J. and Choi, M.S. (2004). Naringin alters the cholesterol biosynthesis and antioxidant enzyme activities in LDL receptor-knockout mice under cholesterol fed condition. Life SCL., 74(13): 1621-34. Kroyer, G. (1986). The antioxidant activity of citrus fruit peels. Z Ernahrungswiss, 25(1): 63-9. Kumar, M.S., Unnikrishnan, M.K, Patra, S., Murthy, K. and Srinivasan, KK. (2003). Naringin and naringenin inhibit nitrite-induced methemoglobin formation. Pharmazie., 58(8): 564-6. Lambev, I., Belcheva, A. and Zhelyazkov, D. (1980a). Flavonoids with antioxidant action (naringin and rutin) and the release of mastocytic and nonmastocytic histamine. Acta Physiol Pharmacol Bulg., 6(2): 70-5. Lambev, I., Krushkov, I., Zheliazkov, D. and Nikolov, N. (1980b). Antiexudative effect of naringin in experimental pulmonary edema and peritonitis. Eksp Med Morfol., 19(4): 207-12. Le Marchand, L., Murphy, S.P., Hankin, J.H., Wilkens, L.R. and Kolonel, L.N. (2000). Intake of flavonoids and lung cancer. J Natl Cancer Inst., 92(2): 154-60. Lee, C.H., Jeong, T.S., Choi, Y.K., Hyun, B.H., Oh, G.T., Kim, E.H., Kim, J.R., Han, J.I. and Bok, S.H. (2001). Anti-atherogenic effect of citrus flavonoids, naringin and naringenin, associated with hepatic ACAT and aortic VCAM-1 and MCP-1 in high cholesterol-fed rabbits. Biochem Biophys Res Commun., 284(3): 681-8. Li, P., Callery, P.S., Gan, L.S. and Balani, S.K (2007). Esterase inhibition by grapefruit juice flavonoids leading to a new drug interaction. Drug Metab Dispos., 35(7): 1203-8. Li, J.M., Che, C.T., Lau, C.B., Leung, P.S. and Cheng, C.H. (2006). Inhibition of intestinal and renal Na+-glucose cotransporter by naringenin. Int J Biochem Cell Biol., 38(5-6): 985-95. Li, F., Meng, F., Xiong, Z., Li, Y., Liu, R. and Liu, H. (2006). Stimulative activity of Drynaria fortunei (Kunze) J. Sm. extracts and two of its flavonoids on the proliferation of osteoblastic like cells. Pharmazie., 61(11): 962-5. Lim, S.C. and Choi, J.S. (2006). Effects of naringin on the pharmacokinetics of intravenous paclitaxel in rats. Bwpharm Drug Dispos., 27(9): 443-7. Martin, M.J., Marhuenda, E., Perez-Guerrero, C. and Franco, J.M. (1994). Antiulcer effect of naringin on gastric lesions induced by ethanol in rats. Pharmacology, 49(3): 144-50. Mata-Bilbao Mde, L., Andres-Lacueva, C., Roura, E., Jauregui, 0., Escribano, E., Torre, C. and Lamuela-Raventos, R.M. (2007). Absorption and pharmacokinetics of grapefruit flavanones in beagles. Br J Nutr., 98(1): 86-92. Middleton, E. Jr. and Drzewiecki, G. (1984). Flavonoid inhibition of human basophil histamine release stimulated by various agents. Biochem Pharmacol., 33(21): 3333-8. Mitsunaga. Y., Takanaga, H., Matsuo, H, Naito, M., Tsuruo, T., Ohtani, H. and Sawada, Y. (2000). Effect of bioflavonoids on vincristine transport across blood-brain barrier. Eur J Pharmacol., 395(3): 193-201. Naderi, G.A., Asgary, S., Sarraf-Zadegan, N. and Shirvany, H. (2003). Anti-oxidant effect of flavonoids on the susceptibility of LDL oxidation. Mol Cell Biochem., 246(1-2): 193-6. Ng, T.B., Ling, J.M., Wang, Z.T., Cai, J.N. and Xu, G.J. (1996). Examination of coumarins, flavonoids and polysaccharopeptide for antibacterial activity. Gen Pharmacol., 27(7): 123740. Ng, T.B., Liu, F. and Wang, Z.T. (2000). Antioxidative activity of natural products from plants. Life ScI., 66(8): 709-23. Orallo, F., Camiiia, M., Alvarez, E., Basaran, H. and Lugnier, C. (2005). Implication of cyclic nucleotide phosphodiesterase inhibition in the vasorelaxant activity of the citrusfruits flavonoid (+I-)-naringenin. Planta Med., 71(2): 99-107. Paredes, A., Alzuru, M., Mendez, J. and Rodriguez-Ortega, M. (2003). Anti-Sindbis activity of flavanones hesperetin and naringenin. Biol Pharm Bull., 26(1): 108-9.
386
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Park, S.H., Park, E.K. and Kim, D.H. (2005). Passive cutaneous anaphylaxis-inhibitory activity of flavanones from Citrus unshiu and Poncirus trifoliata. Planta Med., 71(1): 24-7. Parmar, N.S. (1983). The gastric anti-ulcer activity of naringenin, a specific histidine decarboxylase inhibitor. Int J Tissue React., 5(4): 415-20. Pereira, RM., Andrades, N.E., Paulino, N., Sawaya, A.C., Eberlin, M.N., Marcucci, M.C., Favero G.M., Novak, E.M. and Bydlowski, S.P. (2007). Synthesis and characterization of a metal complex containing naringin and Cu and its antioxidant, antimicrobial, antiinflammatory and tumor cell cytotoxicity. Molecules, 12(7): 1352-66. Rajadurai, M. and Stanely Mainzen Prince, P. (2006). Preventive effect ofnaringin on lipids, lipoproteins and lipid metabolic enzymes in isoproterenol-induced myocardial infarction in Wistar rats. J Biochem Mol Toxicol., 20(4): 191-7. Rajadurai M. and Stanely Mainzen Prince, P. (2006). Preventive effect of naringin on lipid peroxides and antioxidants in isoproterenol-induced cardiotoxicity in Wi star rats: biochemical and histopathological evidences. Toxicology, 228(2-3): 259-68. Rajadurai, M. and Stanely Mainzen Prince, P. (2007). Preventive effect ofnaringin on cardiac markers, electrocardiographic patterns and lysosomal hydrolases in normal and isoproterenol-induced myocardial infarction in Wistar rats. Toxicology, 230(2-3): 178-88. Rajadurai, M. and Prince, P.S. (2007). Preventive effect ofnaringin on isoproterenol-induced cardiotoxicity in Wistar rats: an in VIVO and in vitro study. Toxicology, 232(3): 216-25. Rajadurai, M. and Prince, P.S. (2007). Preventive effect ofnaringin on cardiac mitochondrial enzymes during isoproterenol-induced myocardial infarction in rats: a transmission electron microscopic study. J BlOchem Mol Toxicol., 21(6): 354-6l. Ratty, A.K and Das, N.P. (1988). Effects of flavonoids on nonenzymatic lipid peroxidation: structure-activity relationship. Biochem Med Metab BioI., 39(1): 69-79. Reshef. N., Hayari, Y., Goren, C., Boaz, M., Madar, Z. and Knobler, H. (2005). Antihypertensive effect of sweetie fruit in patients with stage I hypertension. Am J Hypertens., 18(10): 1360-3. Robbins, RC., Martin, F.G. and Roe, J.M. (1988). Ingestion of grapefruit lowers elevated hematocrits in human subjects. Int J Vitam Nutr Res., 58(4): 414-7. Runkel, M., Bourian, M., Tegtmeier, M. and Legrum, W. (1997). The character of inhibition of the metabolism of 1, 2-benzopyrone (coumarin) by grapefruit juice in human. Eur J Clin Pharmacol., 53(3-4): 265-9. Russo, A., Acquaviva, R, Campisi, A., Sorrenti, V., Di Giacomo, C., Virgata, G., Barcellona, M.L. and Vanella, A. (2000). Bioflavonoids as antiradicals, antioxidants and DNA cleavage protectors. Cell BioI Toxicol., 16(2): 91-8. Saponara, S., Testai, L., Iozzi, D., Martinotti, E., Martelli, A., Chericoni, S., Sgaragli, G., Fusi, F. and Calderone, V. (2006). (+I-)-Naringenin as large conductance Ca(2+)-activated K+ (BKCa) channel opener in vascular smooth muscle cells. Br J Pharmacol., 149(8): 1013-2l. Schindler, R and Mentlein, R (2006). Flavonoids and vitamin E reduce the release of the angiogenic peptide vascular endothelial growth factor from human tumor cells. J Nutr., 136(6): 1477-82. Seo, H.J., Jeong, KS., Lee, M.K, Park, Y.B., Jung, U.J., Kim, H.J. and Choi, M.S. (2003). Role of naringin supplement in regulation of lipid and ethanol metabolism in rats. Life Sci., 73(7): 933-46. Shim, C.K, Cheon, E.P., Kang, KW., Seo, KS. and Han, H.K (2007). Inhibition effect of flavonoids on monocarboxylate transporter 1 (MCT1) in Caco-2 cells. J Pharm Pharmacol., 59(11): 1515-9. Shin, Y.W., Bok, S.H., Jeong, T.S., Bae, KH., Jeoung, N.H., Choi, M.s., Lee, S.H. and Park, Y.B. (1999). Hypocholesterolemic effect of naringin associated with hepatic cholesterol regulating enzyme changes in rats. Int J Vitam Nutr Res., 69(5): 341-7. Shiratori, K, Ohgami, K, Ilieva, 1., Jin, X.H., Yoshida, K, Kase, S. and Ohno, S. (2005). The effects of naringin and naringenin on endotoxin-induced uveitis in rats. J Ocul Pharmacol Ther., 21(4): 298-304.
Naringin and its Aglycone, Research and Managements
387
Singh, D., Chander, V. and Chopra, K (2004a). Protective effect of naringin, a bioflavonoid on ferric nitrilotriacetate-induced oxidative renal damage in rat kidney. Toxicology, 201(1-3): 1-8. Singh, D., Chander, V. and Chopra, K (2004b). Protective effect of naringin, a bioflavonoid on glycerol-induced acute renal failure in rat kidney. Toxicology, 201(1-3): 143-51. Singh, D. and Chopra, K. (2004). The effect of naringin, a bioflavonoid on ischemiareperfusion induced renal injury in rats. Pharmacol Res., 50(2): 187-93. So, F.V., Guthrie, N., Chambers, A.F., Moussa, M. and Carroll, KK (1996). Inhibition of human breast cancer cell proliferation and delay of mammary tumorigenesis by flavonoids and citrus juices. Nutr Cancer, 26(2): 167-81. Tsui, V.W., Wong, RW. and Rabie, A.B. (2007). The inhibitory effects of naringin on the growth of periodontal pathogens in vitro. Phytother Res. [Epub ahead of print] Ubeaud, G., Hagenbach, J., Vandenschrieck, S., Jung, L. and Koffel, J.C. (1999). In vitro inhibition of simvastatin metabolism in rat and human liver by naringenin. Life Sci., 65(13): 1403-12. Ueng, Y.F., Chang, Y.L., Oda, Y., Park, S.S., Liao, J.F., Lin, M.F. and Chen, C.F. (1999). In vitro and m vivo effects of naringin on cytochrome P450-dependent monooxygenase in mouse liver. Life Sci., 65(24): 2591-602. Ugocsai, K., Varga, A., Molnar, P., Antus, S. and Molnar, J. (2005). Effects of selected flavonoids and carotenoids on drug accumulation and apoptosis induction in multidrugresistant colon cancer cells expressing MDRlILRP. In vivo, 19(2): 433-8. Wei, M., Yang, Z., Li,P., Zhang, Y. and Sse, W.C. (2007). Anti-osteoporosis activity ofnaringin in the retinoic acid-induced osteoporosis model. Am J Chin Med., 35(4): 663-7. Wenzel, U., Kuntz, S. and Daniel, H. (2001). Flavonoids with epidermal growth factor-receptor tyrosine kinase inhibitory activity stimulate PEPT1-mediated cefixime uptake into human intestinal epithelial cells. J Pharmacol Exp Ther., 299(1): 351-7. Wong, RW. and Rabie, A.B. (2006). Effect of naringin collagen graft on bone formation. Biomaterials, 27(9): 1824-31. Wong, RW. and Rabie, A.B. (2006). Effect of naringin on bone cells. J Orthop Res., 24(11): 2045-50. Wood, N. (2004). The effects of dietary bioflavonoid (rutin, quercetin, and naringin) supplementation on physiological changes in molar crestal alveolar bone-cemento-enamel junction distance in young rats. J Med Food, 7(2): 192-6. Wood, N. (2007). The effects of selected dietary bioflavonoid supplementation on dental caries in young rats fed a high-sucrose diet. J Med Food, 10(4): 694-701. Vanamala, J., Leonardi, T., Patil, B.s., Taddeo, S.S., Murphy, M.E., Pike, L.M., Chapkin, RS., Lupton, J.R and Turner, N.D. (2006). Suppression of colon carcinogenesis by bioactive compounds in grapefruit. Carcinogenesis, 27(6): 1257-65. Yeh, S.L., Wang, W.Y., Huang, C.H. and Hu, M.L. (2005). Pro-oxidative effect of beta-carotene and the interaction with flavonoids on UVA-induced DNA strand breaks in mouse fibroblast C3H10T1/2 cells. J Nutr Biochem., 16(12): 729-35. Yeum, C.H. and Choi, J.S. (2006). Effect of naringin pretreatment on bioavailability of verapamil in rabbits. Arch Pharm Res., 29(1): 102-7. Younes, M. and Siegers, C.P. (1981). Inhibitory action of some flavonoids on enhanced spontaneous lipid peroxidation following glutathione depletion. Planta, 43(11): 240-4. Yu, J., Wang, L., Walzem, RL., Miller, E.G., Pike, L.M. and Patil, B.S. (2005). Antioxidant activity of citrus limonoids, flavonoids, and coumarins. J Agric Food Chem., 53(6): 200914.
"This page is Intentionally Left Blank"
19 Biotransformation of Drugs
ABSTRACT Biotransformation is the termination or alteration of biologic activity of a drug or xenobiotic. In general, lipophilic drugs are transformed to more polar and hence readily excretable products. Most metabolic biotransformations occur at a point between absorption of a drug into general circulation and its renal elimination. The major sites for biotransformation are liver, gastrointestinal tract, the lungs, skin and the kidneys. The enzymes involved in the metabolism are either microsomal or nonmicrosomal. In general all the reactions can be assigned to two major categories called phase I and phase II. The phase lor non synthetic reactions involve oxidation, reduction, hydrolysis, cyclization and decyclization. The phase II or synthetic reactions involve glucuronide conjugation, acetylation, methylation, sulfate conjugation, glycine conjugation, glutathione conjugation and ribonucleoside/nucleotide synthesis. Certain drugs are inactivated spontaneously e.g. the Hoffman elimination of atracurium. Some drugs like the lithium and streptomycin do not under go any metabolic transformation. Key words : Xenobiotic, metabolism, conjugation, hydrolysis, renal, synthetic, reaction
INTRODUCTION Living organisms metabolize drugs and other xenobiotics through a natural process that involves the same enzymatic processes that are utilized for metabolism of dietary substances (Jenner & Testa, 1981). The capacity to metabolize unusual xenobiotics and other food sources is important for 1. Department of Pharmacology, Christian Medical College, Vellore - 632 002, Tamil Nadu, India. * Corresponding author : E-mail: [email protected]
390
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
survival of living beings. Drugs are considered as xenobiotics and most of them undergo extensive metabolism in humans. A large plethora of diverse enzymes have evolved in animals with the sole function of metabolizing foreign chemicals. Enzymes that metabolize xenobiotics have been called as drug-metabolizing enzymes, though they are involved in the metabolism of many foreign chemicals to which humans are exposed (Correia, 2006). For example, lipophilic barbiturates such as thiopental and pentobarbital would have extremely long half lives if it were not for their metabolic conversion to water soluble compounds. The lipophilic nature of renal tubular membranes also facilitates the reabsorption of hydrophobic compounds following their glomerular filtration. Consequently, most drugs would have a prolonged duration of action if termination of their action depended solely on renal excretion. Lastly, drug metabolizing enzymes have been exploited through the design of pharmacologically non active prodrugs that are converted in vivo to pharmacologically active drugs.
SITES OF DRUG METABOLISM Drug metabolizing enzymes are found in most of the tissues in the body with highest levels in liver and the gut. The liver is considered as the major site for degradation of endogenous chemicals for e.g. cholesterol and steroid hormones. Other organs that contain significant xenobiotic - metabolizing enzymes include the kidneys and lung and nasal mucosa, the latter two playing significant roles in first-pass metabolism of drugs that are administered through aerosol sprays. Other tissues that display considerable activity include skin. Although drug metabolism within a living body can occur by spontaneous, uncatalyzed chemical reactions, the majority of transformations are catalyzed by specific cellular enzymes (Gonzalez & Tukey, 2006). At the subcellular level these enzymes may be situated in the mitochondria, endoplasmic reticulum, cytosol, lysosomes or even the nuclear envelope or plasma membrane.
PHASES OF DRUG BIOTRANSFORMATION • Biochemical alteration of the drug in the body • It is needed to render non polar (lipid soluble) compounds, to polar (lipid insoluble) so that they are not reabsorbed in the renal tubules
• Hydrophilic drugs e.g. streptomycin, Neostigmine, decamethonium etc. are not biotransformed
Biotransformation of Drugs
391
CONSEQUENCES OF BIOTRANSFORMATION Active drug to inactive metabolite (inactivation)
Active drug to active metabolite
e.g. Morphine, chloramphenicol, pentobarbitone, propranolol
Inactive drug to active metabolite (prodrug)
e.g. Codeine - Morphine Phenacetin - Paracetamol - Digoxin Digitoxin
e.g. Levodopa Enalapril a-methyldopa
Diazepam Desmethyl Amitriptyline
Prednisone
- Oxazepam - Diazepam - Nortriptyline
Bacampicillin Sulfasalazine
- Dopamine - Enalaprilat - a methyl norepinophrine - Prednisolone - Ampicillin - 5 - Amino salicylic acid
PHASES IN METABOLISM OF DRUGS The chemical reaction of biotransformation can take place in two phases 1. Phase IINon - synthetic reactions
- Metabolite may be active or inactive 2. Phase II1Synthetic reactions - Metabolite is mostly inactive Unchanged ~
Drug
~
Excretion
~. frusemide, atenolol
Phase I
1
Metabolite
~ Excretion
Phase II
~ Metabolite
~
Excretion
Note: If the metabolite of phase I is not sufficiently polar to be excreted, it undergoes phase II reactions
392
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Phase I Reactions Non-Synthetic Reactions - Phase I reactions convert the drug to a more polar metabolite by oxidation, reduction, hydrolysis, cyclization and decyclization - Phase I reactions introduce or expose a functional group on the parent compound - The enzyme systems involved in phase I reactions are located primarily in endoplasmic reticulum (phase II conjugation enzymes are mainly cytosolic)
OXIDATION Oxidation is the process of addition of oxygen (or a negatively charged radical) to a drug molecule or removal of hydrogen (or a positively charged radical) from a drug molecule. Oxidation reactions are the most of monooxygenases in the liver, which in the final step involve a cytochrome P-450 haemoprotein, NADPH, cytochrome P-450 reductase and O2 (Ortiz de Montellano & Corriea, 1995). More than 100 cytochrome P-450 isoenzyme differing in their affinity for various substrates (drugs) have been identified. The cytochrome P-450 isoenzymes important in human beings are given as following: 1) CYP3A4/5
• Carryout biotransformation of largest number of drugs (nearly 50%) present in liver and also expressed in intestine and kidney. This isoenzyme is inhibited by erythromycin, clarithromycin, ketoconazole, itraconazole, verapamil, diltiazem, ritonavir, a constituent of grape fruit juice - induced by - rifampicin, barbiturates, anticonvulsants (Thummel & Wilkinson, 1998) 2)CYP2D6
• Next most important CYP isoform • Metabolizes nearly 20% of drugs, including tricyclic antidepressants, selective serotonin receptake inhibitors, neuroleptics, antiarrhythmics, ~-blockers and opiates (Guengrich, 1997) • Isoenzyme inhibited by quinidine, results in failure of conversion of codeine to morphine analgesic effect of codeine is lost
Biotransformation of Drugs
393
3)CYP2C819
• >15% of drugs are biotransformed by this isoenzyme • Drugs metabolized include - phenytoin and warfarin 4)CYP2C19
• > 12% of drug
• Drugs metabolized include omeprazole, lansoprazole • Isoenyzme induced by rifampicin and carbamazepine 5)CYPIA1I2
• Participates in the metabolism of only few drugs • This isoenzyme is important for activation of procarcinogens • Inducers include (1) Rifampicin
(2) Carbamazepine (3) Polycyclic hydrocarbons (4) Cigarette smoke (5) Smoked meat (charbroiled meat) 6)CYP2El
• Catalyses formation of minor metabolites of few drugs, especially the hepatotoxic N -acetyl benzoquinonemine from paracetamol • Chronic alcoholism induces the isoenzyme The relative amount of different cytochrome P-450 differs among species and among individuals ofthe same species. These differences largely account for marked interspecies and inter individual differences in rate of metabolism of drugs Various oxidation reactions are 1. N-dealkylation 2. O-dealkylation 3. Aliphatic hydroxylation 4. Aromatic hydroxylation 5. N-oxidation 6. S-oxidation 7. Deamination
394
Compo Bio. Nut. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
S.No.
Oxidative reactions
1.
N-dealkylation
RNCH3~RNH2+
CH20
Imipramine, diazepam, codeine, erythromycin, morphine, tamoxifen, theophylline, caffeine
2.
O-dealkylation
ROCH 3 ~ ROH + CH 20
Codeine, indomethacin, dextromethorphan
3.
Aliphatic hydroxylation
Reaction
Examples
OR
I
RCH 2CH 3 ~ R-CRCH 3 R
4.
Aromatic hydroxylation
@-Q ~
Tolbutamide, ibuprofen, pentobarbital, meprobamate, cyclosporine, midazolam Phenytoin, phenobarbital, propranolol, phenylbutazone, ethinylestradiol, amphetamine, warfarin
OH
5.
N-oxidation
RNH2~RNHOH
R 1"/NH~ R 1"/N-OH R2 R2 6.
8-oxidation
7.
Deamination
R 1"/8 ~ R 1"/8=0 R2 R2 OH
Chlorpheniramine, dapsone, meperidine, quinidine, acetaminophen Cimetidine, chlorpromazine, thioridazine, omeprazole Diazepam, amphetamine
I I
RCHCH 3 ~ R-C-CH3
I
NH2
NH2 0
~
II
R-C-CH3 + NH2
REDUCTION This reaction is the converse of oxidation and involves cytochrome P-450 enzymes working in opposite directions. Drugs primarily reduced are chloralhydrate, chloramphenicol, halothane. There are many types of reduction reactions. 1. Nitro reduction - e.g. chloramphenicol -Arylamine
2. Keto reduction - e.g. cortisone - hydrocortisone
Biotransformation of Drugs
395
Note: Reduction may be catalysed by non microsomal enzymes also. e.g. disulfiram and nitrates HYDROLYSIS This is the cleavage of drug molecule by taking up a molecule of water. Ester + H 2 0
Esterase
~
.
ACId + Alcohol
Similarly amides and polypeptides are hydrolysed by amidases and peptidases. Hydrolysis occurs in liver, intestine, plasma and other tissues. Hydrolysis reactions Reaction (i)
o II
R 1COR2 ~ R1COOH + R 20H (ii)
o II
R 1COR2 ~ R1COOH + R2NH2
Example Procaine, aspirin, clofibrate, meperidine, enalapril, cocaine Lidocaine, procainamide, indomethacin
CYCLIZATION This is formation of ring structure from a straight chain compound e.g. proguanil DECYCLIZATION This is the opening up of ring structure ofthe cyclic drug molecule. This is a minor pathway e.g. barbiturates, phenytoin
Phase II Reactions Phase II reactions are carried out by enzymes that are synthetic in nature. The metabolites of phase I reaction are conjugated to endogenous substances in phase II reaction. These endogenous substances are usually derived from carbohydrate or aminoacid source. The formation of conjugate involves highenergy intermediates and specific transfer enzymes, which requires high energy. Such transferase enzymes are either located in micro somes or in the cytosol. This results in termination of biological activity of the drug. However, certain conjugation reaction (i.e. acyl glucuronidation ofNSAIDS, N-acetylation ofINH) may lead to formation of reactive species responsible for the hepatotoxicity of the drug. The various phase II reactions include, glucuronidation, acetylation, methylation, sulfate conjugation, glycine conjugation and glutathione conjugation.
396
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
Glucuronidation This reaction is catalysed by UDP-Glucuronic transferases (UGTs). It is one of the most important reactions in synthetic reactions. These enzymes catalyse the transfer of glucuronic acid from the co-factor UDP - glucuronic acid. Compunds with hydroxyl or carboxylic acid group are easily conjugated with glucuronic acid e.g. aspirin, acetaminophen, morphine, digoxin (Kroemer & Klotz, 1992; Dutton, 1980). Some of the endogenous substrates including, bilirubin, steroid hormones and thyroxine utilizes this pathway for its met~bolism. The UGTs are present in high concentrations in specific tissues like GI tract and liver. Glucuronides are either excreted by the kidneys into the urine or through active transport ofliver hepatocytes into the bile. These acids are re-absorbed to the liver by entero-hepatic re-circulation. Many drugs metabolized by liver utilizes the same process and re-enter circulation. The commensals located in the lower GI tract releases P-D-glucopyrano-siduronic acid and liberates the free drug into the intestinal lumen. Similar to water reabsorption, the drug also gets absorbed passively from the intestinal lumen and re-enter circulation.
Acetylation N-acetyl transferases (NATs) are located in the cytosol and they are involved in addition of acetyl group from the co-factor acetyl coenzyme A. Compounds having amino or hydrazine residues undergoes acetylation e.g. Dapsone, sufonamides, clonazepam, INH (Vatsis et al., 1995). The N-acetyl tranferases in human shows extreme genetic polymorphism ranging from fast or rapid acetylators to slow acetylators. The administration of therapeutic dose of drugs like hydralazine or INH leads to toxicity of the drug. Slow acetylation are also predisposed to idiosyncratic hypersensitivity reactions and drug-induced auto-immune disorders.
Methylation Some xenobiotics and drugs undergo 0-, N - and S-methylation. Methionine and cystiene acts as methyl donor. Commonly the amines and phenols undergo methylation is carried out by group of enzymes known as N -methyl tranferases (Weinshilboum et al., 1999). They are catechol-O-methyl transferase (COMT), phenolromethyl transferase (POMT), a thiopurine-Smethyl transferase (TPMT) and thiol-methyl transferase includes TMT, which catalyses S-methylation of azathioprine, 6-mercaptopurine and thioguamine. Dopamine, epinephrine, histamine and thiouracil are some of the drugs which undergo methylation.
Biotransformation of Drugs
397
Sulfate conjugation Aromatic and aliphatic compounds are acted upon by sulfotransferases (SULTS) (Glatt, 2000). These enzymes are located in the cytosol and conjugate sulfate to hydroxyl groups of aromatic and aliphatic compounds. SULTS are responsible for wide variety of xenobiotic metabolism and many isoforms ofthis enzyme have been identified (Negishi et al. , 2001). The drugs which undergo sulfation include chloromphenicol, adrenal and sex steroids. SULTs also play an important role in normal human homeostasis. e.g. SULT (B) is expressed in skin and brain and carries out catalysis of cholesterol and thyroid hormones. A significant proportion of catecholamines and sex hormones exist in sulfated form, due to the action ofSULT.
Glycine conjugation This reaction is carried out by Acyl-CoA glycine tranferase. These enzymes are located in the mitochondria of hepatocyte. Salicylic acid, benzoic acid, nicotinic acid and cinnamic acids undergo glycine conjugation, although this is not a major pathway of metabolism.
Glutathione conjugation Glutathione-S-tranferase are located in the microsome and cytosol (Hayes et al., 2004). This enzyme catalyzes the transfer of glutathione to reactive electrophiles. Glutathione exist either as oxidised form (GSSG) or as reduced form (GSH) in the cell (Townsend & Tew, 2003). The GSH: GSSG ratio is important for maintaining cellular environment in reduced state. A reduced intracellular level ofGSH can lead to severe oxidative stress and damage to the cell. In this pathway a mercapturate is formed, which serves to inactivate highly reactive quinone or epoxide intermediate formed during metabolism of certain drugs e.g. paracetamol. In condition like paracetamol overdose, large amounts of quinines are formed. This leads to reduced level of intracellular glutathione, which causes cell damage. However, this can be presented by supplying enogenous glutathione in the form of N-acetyl cysteine. Almost all xenobiotics are metabolized by many pathways, either simultaneously or sequentially. As a result one drug can produce variety of metabolites. However, enzymes of intermediary metabolism like alcohol dehydrogenase, xanthine onidase, plasma choline esterase and monoamine oxidase also metabolise compounds like alcohol, allopurinol, succinylcholine and adrenaline respectively. Some drugs undergo spontaneous molecular re-arrangement without any enzyme. Classical example is metabolism of atracurium and this is called as Hofmann elimination (Zheng et al., 1997).
398
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
REFERENCES Correia, M.A. (2001). Drug biotransformation In: Basic & Clinical pharmacology Ed. B.G. Katzung-Bth Edition, Page 51-63. Dutton, G.J. (1980). Glucuronidation of drugs and other compounds. CRC Press, Inc., Boca Raton. Glatt, H. (2000). Sulfotransferases in the bioactivation of xenobiotics. Chem BioI. Interact., 129: 141-170. Gonzalez, F.J. and Tukey, RH. (2006). Drug metabolism. Chapter 3, 11th Edition Guengrich, F.P. (1997). Role of cytochrome P450 enzymes in drug-drug interactions. Adv. Pharmacol., 43: 7. Hayes, J.D., Flanagan, J.U. and Jowsey, LR (2004). Glutathione transferases. Annu. Rev. Pharmacol. Toxicol. Jenner, P. and Testa, B. (Editors) (1981). Concepts in Drug metabolism. Part B of: Drugs & pharmaceutical science series, Vol.lO, Marcel Dekker. Kroemer, H.K. and Klotz, U. (1992l. Glucurodination of drugs: A reevaluation of the pharmacological significance of conjugates and modulating factors. Clin. Pharmacokinet., 23: 292. Negishi, M., Pederson, L.G. and Petrochenko, E. (2001). Sructure and function of sulfotransferases. Arch Biochem Biophys., 390: 149-157. Ortiz de Montellano, P.R and Correia, M.A. (1995). Inhibition of cytochrome P450 enzymes. In: Cytochrome P450: Structure, Mechanism & Biochemistry, 2nd Edn. Ortiz de Montellano P. (Ed.) Plenum Press. Thummel, KE. and Wilkinson, G.R (1998). In vitro and in vivo drug interactions involving human CYF3A. Ann. Rev. Pharmacol. Toxicol., 38: 389. Townsend, D.M. and Tew, KD. (2003). The role of glutathione s transferase in anti-cancer drug resistance. Oncogene, 22: 7369-7373. Vatsis, KP., Weber, W.W. and Bell, D.A. (1995). Nomenclature for N-acetyltransferases. Pharmacogenetics, 5: 1-17. Weinshilboum, R.M., Otterness, D.M. and Szumlanski, C.L. (1999). Methylation pharmacogenetics, catechol o-methyl transferase, thiopurine methyl transferase and histamine N-methyltransferase. Annu. Rev. Pharmacol. Toxicol., 39: 19-52. Zheng, H., Lin, Y. and Chen, S. (1997). Application of atracurium to critical care patients in ICU. Zhonghua wai ke za zhi., 35(2): 125-126.
20 Pharmacokinetics and Drug Interactions of GlycyrrhiziniGlycyrrhiza
ABSTRACT Glycyrrhiza glabra (licorice) root contains a triterpenoid saponin called glycyrrhizin. Glycyrrhizin is being used in the treatment of hepatitis. Pharmacokinetics of glycyrrhizin has been evaluated in rats following intravenous (iv) and oral administration. Glycyrrhizin is a glycoside containing 2 glucuronic acid molecules attached to glycyrrhetic acid, the aglycone. After intravenous administration in rats, glycyrrhizin is secreted from liver into the bile and comes to intestine where it is hydrolyzed by intestinal bacteria to glycyrrhetic acid which is absorbed into the systemic circulation. Glycyrrhetic acid also is secreted from the liver as unchanged glycyrrhetic acid and glycyrrhetic acid monoglucoronide which is hydrolyzed in the intestine, reabsorbed and undergoes enterohepatic cycling thereby reducing clearance of the drug and prolonging its residence in the body. Thus decline in plasma concentration of glycyrrhizin, following iv administration is biexponential. Since secretion is an active process involving carriers, the transport of drug from liver to bile is a saturable process and pharmacokinetics of glycyrrhizin is nonlinear. Glycyrrhizin showed a dose-dependent pharmacokinetics. Glycyrrhizin and glycyrrhetic acid both being actively secreted and having molecular weight >250, would have high biliary clearance. Oral absorption of glycyrrhizin is very poor (bioavailability 1%), it is slowly metabolized by the intestinal bacteria to
1. Delhi Institute of Pharmaceutical Sciences and Research, Formerly College of
*
Pharmacy, (University of Delhil, Pushp Vihar, Sector III, New Delhi - 110017, India. Corresponding author: E-mail: dkmajumdaa:[email protected];[email protected]
400
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
glycyrrhetic acid and absorbed. Intraperitoneal and nasal administration in rats has been found to improve bioavailability of glycyrrhizin. Studies in human subjects indicate oral glycyrrhizin is partly absorbed from gastrointestinal tract in intact form; rest of the dose is metabolized to glycyrrhetic acid and absorbed. The decline in plasma concentration following iv administration of glycyrrhizin is biexponential and no dose dependency of drug disposition has been observed. Glycyrrhizin iv administration in cirrhosis/hepatitis patients showed a monoexponential decline of drug in plasma indicating a linear pharmacokinetics of the drug and the pharmacokinetics are closely related to the extent of liver function. Similarly pharmacokinetics of glycyrrhizin (iv), in patients with chronic hepatitis C infection, has also been linear but no correlation between hepatic function and pharmacokinetics is observed which may be explained by the patients having milder liver disease. Oral administration of amoxicillin and metronidazole could destroy intestinal bacteria and thereby make coadministered glycyrrhizin ineffective. Glycyrrhizin could inhibit hepatic metabolism of corticosteroids like prednisolone resulting in toxicity. Licorice (glycyrrhizin) if concurrently administered with anticoagulant could cause bleeding due to its antiplatelet / antithrombin effect. Concurrent administration of licorice with potassium-losing diuretics, laxatives cause excessive potassium loss and may need potassium supplement. Licorice induced hypokalemia may cause digoxin toxicity. Licorice in combination with contraceptive may cause sodium and fluid retention and elevated blood pressure. Key words: AVC, CLtotal, enterohepatic, exponential, glycyrrhizin, glycyrrhetic acid, MRT, hypokalemia
INTRODUCTION Glycyrrhiza glabra (Leguminosae) is a plant of about 1.5 meter height having purplish blue flower. The dried roots and stolons of the plant are used as drug. Glycyrrhiza is also known as licorice root. Glycyrrhiza contains glycyrrhizin, a triterpenoid saponin and its sweet principle, and other bioactive components e.g. saponins, flavones, isoflavones, coumarins, triterpene sterols etc. Glycyrrhizin is a glycoside of glycyrrhetic (glycyrrhetinic) acid (Fig 1). On hydrolysis the glycoside loses its sweet taste and is converted to the aglycone glycyrrhetic acid and two molecules of glucuronic acid (Vyas & Majumdar, 2005).
Pharmacokinetics and Drug Interactions of Glycyrrhizin / Glycyrrhiza
401
COOH
E~,,-o
o~~
aglYCon:: Glycyrrhetinic acid/Glycyrrhetic acid
E~J
o~ OH glycone
=2 molecules of glucuronic acid GlycyrrhiziniGlycyrrhizinic acid/Glycyrrhizic acid
Fig 1. Glycyrrhizin
IUPACNAME (3- ~,20- ~)-20-Carboxy-11-oxo-30-norolean-12-en-3-yl glucopyranuronosyl-a-D-glucopyranosiduronic acid.
2-0- ~-D
Glycyrrhizin (glycyrrhizic acid, glycyrrhizinic acid, glycyrrhetinic acid glycoside) (C 42 H62 016)' mol weight 822.93, C=61.30%, H=7.59%, 0=31.11 %, has been crystallized from glacial acetic acid. It has intensely sweet taste. It is freely soluble in hot water, alcohol and practically insoluble in ether. Ammonium glycyrrhizinate pentahydrate (C 42 H65 N0 16 . 5 H 20) has been crystallized as needles from 75% aqueous ethanol. The salt melts around 212-217 degree Celsius with decomposition. It is soluble in ammonia water and glacial acetic acid. It has UV absorption maximum at 248 nm. Dipotassium salt of glycyrrhizinic acid (C 42 H60 ~ 016)' mol wt 899.11 has also been reported (Merck Index, 2006).
USES Traditionally licorice is used as demulcent and expectorant. The liquid extract is used in cough mixtures and as a flavoring agent in mixtures containing nauseous medicines e.g. ammonium chloride, the alkali iodide, quinine and cascara liquid extract. A decoction with linseed has been used as a domestic treatment of cough and bronchitis. Due to its mild antiinflammatory and mineralocorticoid activities, licorice has been used as a substitute to corticosteroids. Deglycyrrizinated licorice having reduced mineralocorticoid activity has been used in peptic ulcer (Reynolds, 1989).
402
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Recent studies indicate anti-inflammatory, antiallergic, antioxidant, antithrombin, antiviral, hepatoprotective, immunomodulatory, and anti tumour activities of glycyrrhizin, an active constituent of Glycyrrhiza glabra (Vyas & Majumdar, 2005). In Japan and India glycyrrhizin is being used in the treatment of hepatitis. Injection and tablet formulations of mono ammonium glycyrrhizinate are available in the market. Studies in the recent past have helped in better understanding of the metabolism/pharmacokinetics and adverse reaction of glycyrrhizin and glycyrrhiza. The relevant articles has been retrieved from the Medline and presented in the following section.
METABOLISM AND PHARMACOKINETICS Ichikawa et al. (1986) studied biliary excretion of glycyrrhizin using rat with or without biliary fistulization. The plasma decay in the control rats without fistulization following an intravenous (iv) dose of 100 mg/kg of glycyrrhizin was generally biphasic. However, secondary peaks were observed in all rats in the elimination phase i.e. 0.5 to 12 h following dosing. The plasma concentration in the rats with biliary fistulization administered same dose showed a biexponential decline. The area under the plasma concentration-time curve (AVC) and total clearance (CLtotal) were significantly higher and lower in control rats respectively. The biliary excretion was 80.6 ± 9.9% ofthe administered dose and intestinal absorption was confirmed by using the bile collected after iv dosing. It was concluded that the glycyrrhizin was predominantly secreted from the liver into the bile and that the secondary peak in the elimination phase, the higher AVC and the lower total clearance in the control rats were due to the effects of the enterohepatic cycling of glycyrrhizin. Furthermore the transport of the drug from liver to the bile appears to be a saturable process For appreciable biliary excretion the drug should be polar. It should be actively secreted and its molecular weight should be more than 250. Glycyrrhizin is actively secreted in the bile and its molecular weight is around 800. Thus it seems likely that glycyrrhizin will have high biliary clearance. In a subsequent study pharmacokinetics of glycyrrhetic acid, a major metabolite of glycyrrhizin was evaluated in rats after bolus iv injection at a dose of 2, 5 or 12 mg/kg. The decline in plasma concentration was generally biexponential at each dose but the terminal disposition became much slower with increase in dose. A greater than proportional increase in plasma glycyrrhetic acid concentration was observed with increase of dose, suggesting a dose dependent glycyrrhetic acid disposition. Apparent total body clearance decreased significantly, with increase of dose. On the other
Pharmacokinetics and Drug Interactions of Glycyrrhizin I Glycyrrhiza
403
hand apparent steady state distribution volume after iv administration was unaffected by the dose. The plasma disposition at each dose fitted well to a two compartment pharmacokinetic model with Michaelis-Menten elimination. It was concluded that the pharmacokinetics of glycyrrhetic acid in the rat is dose dependent owing to a saturable elimination rate. The plasma level of glycyrrhetic acid after iv dosing (100 mg/kg) of glycyrrhizin in the control rats (without biliary fistulation) sustained the concentration range of 1.5-3 mglmL during 1-48 h but that in the rats with biliary fistulation declined with time. It was suggested that the sustained plasma level of glycyrrhetic acid is accounted for by the intestinal absorption of glycyrrhetic acid produced from glycyrrhizin and glycyrrhetic acid conjugates during the enterohepatic cycling of both (Ishida et al., 1989). Tsai et al. (1992) studied pharmacokinetics of glycyrrhizin in rat after bolus iv administration at a dose of 20, 50 or 100 mg/kg. Concentration of glycyrrhizin was measured by HPLC. The decline in the concentration of glycyrrhizin in plasma was generally biexponential at each dose but the terminal disposition became much slower as the dose was increased. A greater than proportional increase in the glycyrrhizin concentration in plasma was observed with an increase in the dose, a result suggesting dose-dependent glycyrrhizin disposition. The disposition of drug in plasma at each dose level fitted well to a two compartment pharmacokinetic model. The apparent total body clearance decreased significantly with increase in the dose. On the other hand the apparent volume of distribution after iv administration was unaffected by the dose. The results indicate that the pharmacokinetics of glycyrrhizin is nonlinear. Ishida et al. (1992) also investigated dose dependent pharmacokinetics of glycyrrhizin by measuring drug disappearance from plasma and biliary excretion in rats. The decline in plasma concentration was biexponential after iv dose of 5, 10, 20 or 50 mg/kg. Dosage however, had a marked effect on the pharmacokinetics with greater than proportional increase in area under the plasma concentration curve (AUC) at doses 20 and 50 mg/kg, even though the increase was proportional at doses of 5 and 10 mg/kg. There was also a significant increase of the steady state distribution volume (Vdss) as well as significant decreases in total body clearance (CL total) and biliary clearance at 20 and 50 mg/kg from those at 5-10 mg/kg and 5-20 mg/kgrespectively. The AUC, Vdss and renal clearance (CLR) at a given dose showed no significant difference between rats with or without bile fistulas. The plasma unbound fraction (0.006-0.026) increased with increasing plasma glycyrrhizin concentration over the observed range (2-900 microgram/mL). No significant difference in Vdss for unbound glycyrrhizin was observed between the doses which indicate that the distribution of glycyrrhizin into tissues is not changed by an increase of dose. On the other hand a dose dependency of CL total for unbound glycyrrhizin was observed and confirmed to be attributable to
404
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
dose dependency in CLB for unbound glycyrrhizin since there was no significant difference in CLR or metabolic clearance for unbound glycyrrhizin between the doses. In another study the role of enterohepatic circulation of glycyrrhetic acid in rats, was determined by kinetic analysis of glycyrrhetic acid. The concentration of glycyrrhetic acid in the plasma of control rat (without bile duct cannulation) during the first 5 h after iv administration of glycyrrhetic acid (2, 5, 10 and 20 mg/kg) were similar to those in the bile duct cannulated rat at each dose. No significant difference was observed in the values of the terminal half lives, total body clearance, the distribution volume at steady state, and the area under the curve of concentration in plasma versus time and the mean residence time (MRT) in each dose between both groups. When glycyrrhetic acid (2, 5, 10 and 20 mglkg) was administrated iv to the bile duct cannulated rat, excretion of unchanged glycyrrhetic acid in bile was <1% of each dose, and that of glycyrrhetic acid 3-0-glucuronide was 12%. In the control rat, a secondary peak of glycyrrhetic acid was observed 12 h after iv administration of glycyrrhetic acid (20 mg/kg). The enterohepatic circulation of glycyrrhetic acid was confirmed by the linkrat method in which bile of the donor rat after iv administration of glycyrrhetic acid (20 mg/kg) was allowed to flow directly into the duodenum of the recipient rat. Glycyrrhetic acid was found in the plasma of the recipient rat after 6 h and its concentration reached maximum (approximately 0.5 mg!mL) 8-12 h after dosing the donor rat (Kawakami et al., 1993). Akao et al. (1994) prepared gnotobiote rats by infecting germ free rats with Eubacterium sp. strain GLH, a human bacterium capable of hydrolyzing glycyrrhizin to 18-beta-glycyrrhetic acid. Glycyrrhizin (100 mg! kg) was administered orally to conventional, germ free and gnotobiote rats but no glycyrrhizin could be detected in plasma 4 or 17 h after the administration. Plasma 18-beta-glycyrrhetic acid was not detected 4 or 17 h neither after the administration of glycyrrhizin to germ free rats nor could this compound be detected in caecal contents or in the faeces. However 18-beta-glycyrrhetic acid (0.6-2.6 nmole/mL) was detected in the plasma of the conventional and gnotobiote rats 4 and 17 h after the administration and the caecal contents after 4 h and the cumulative faeces up to 17 h of the conventional and the gnotobiote rats contained considerable amount of 18-beta glycyrrhetic acid. These findings indicate that orally administered glycyrrhizin is poorly absorbed from the gut but is hydrolyzed to 18-betaglycyrrhetic acid by intestinal bacteria such as Eubacterium sp. strain GLH and the resulting 18-beta-glycyrrhetic acid is absorbed. To clarifY the metabolic fate of glycyrrhizin when orally ingested, a subsequent study investigated the bioavailability of glycyrrhetic acid after intravenous or oral administration of glycyrrhetic acid (5.7 mglkg equimolar
Pharmacokinetics and Drug Interactions of Glycyrrhizin I Glycyrrhiza
405
to glycyrrhizin) or glycyrrhizin (10 mg/kg) at a therapeutic dose in rats. Plasma concentration of glycyrrhetic acid rapidly declined after its iv administration with AUe of 9200 ± 1050 ng.hlmL and MRT of 1.1 ± 0.2 h. The AUe and MRT values after oral administration were 10600± 1090 ng.h/mL and 9.3 ± 0.6 h respectively. After oral administration of glycyrrhizin the parent compound was not detectable in plasma at any time but glycyrrhetic acid was detected at a considerable concentration with AUe of 11700 ± 1580 ng.hlmL and MRT of 19.9 ± 1.3 h while glycyrrhetic acid was not detected in the plasma of germ free rats at 12 h after oral administration of glycyrrhizin. The AUe value of glycyrrhetic acid after oral administration of glycyrrhizin was comparable with those after intravenous and oral administration of glycyrrhetic acid indicating a complete biotransformation of glycyrrhizin to glycyrrhetic acid by intestinal bacteria and a complete absorption of the resulting glycyrrhetic acid from the intestine. Plasma glycyrrhizin rapidly decreased and disappeared in 2h after intravenous administration. AUe and MRT values were 2410 ± 125 microgram minlmL and 29.8 ± 0.5 min respectively. Plasma concentration of glycyrrhetic acid showed two peaks, a small peak at 30 min and a large peak at 11.4 h after intravenous administration of glycyrrhizin, with an Aue of 15400 ± 2620 ng.hlmL and an MRT of 18.8 ± 1.0 h. The plasma concentration profile of the later large peak was similar to that of glycyrrhetic acid after oral administration of glycyrrhizin, which slowly appeared and declined. The difference ofMRT values (19.9 and 9.3 h) for plasma glycyrrhetic acid after oral administration of glycyrrhizin and glycyrrhetic acid suggests slow conversion of glycyrrhizin to glycyrrhetic acid in the intestine (Takeda et al., 1996). Pharmacokinetic behavior of glycyrrhizin after intravenous (iv), oral and intraperitoneal (ip) administration was compared in rats. The elimination half life, total body clearance and volume of distribution at steady state of glycyrrhizin were not significantly different among doses (2, 10, 50 mg/kg. iv).Glycyrrhizin was only detected in plasma (maximum level 1.3 mg/mL) after oral administration of 50 mg/kg. From comparison of the area under the plasma concentration-time curve after iv and oral administration of 50 mg/kg, the bioavailability of glycyrrhizin was estimated to be approximately 1%. Glycyrrhizin was stable for at least 3h in gastric juice. The plasma concentration of glycyrrhizin after oral administration to neomycin treated rats was not significantly different from that after administration to untreated rats. From the results it appeared that the extremely low bioavailability by the oral route may be due to poor absorption of glycyrrhizin from the intestinal tract. On the other hand, the plasma concentration of glycyrrhizin rapidly increased after ip administration of doses of 2, 10 and 50 mg/kg and reached a maximum level (4.7, 33, 238.9 microgramlmL respectively) with in 30 min. The bioavailability (65-90%) of glycyrrhizin after ip administration was enhanced dramatically. The ip route of
406
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
administration may thus improve the bioavailability of glycyrrhizin (Yamamura et al., 1995). A recent study has reported improved bioavailability of glycyrrhizin in rat after nasal administration. The absolute availability has been reported to be 20% which was much higher than that observed after oral administration (Sasaki et al., 2003). Pharmacokinetic profile of glycyrrhizin and its metabolites after oral and intravenous administration of glycyrrhizin to healthy human volunteers has also been reported. An improved HPLC method was used to monitor glycyrrhizin and its metabolites in plasma, and glycyrrhizin could be detected at 500 ng/mL level. After oral administration of glycyrrhizin (100 mg) to three normal subjects, the major metabolite of glycyrrhizin (i.e. glycyrrhetic acid) appeared in plasma «200 ng/mL) but glycyrrhizin was not found. On the other hand glycyrrhizin was found in urine and the amount excreted was 1.1-2.5% of the dose. This finding suggests that glycyrrhizin is partly absorbed in the intact form from the gastro-intestinal tract. The concentration of glycyrrhizin in plasma after iv administration of glycyrrhizin (40,80, 120 mg) showed biexponential profile during the 24 h period after administration of each dose, which suggests a two compartment model of distribution of the drug. The glycyrrhizin metabolite, glycyrrhetic acid and glycyrrhetic acid -3-0-glucuronide were not detected in either plasma or urine. The terminal half life of glycyrrhizin, the apparent volume of central compartment, the steady state distribution volume and the total body clearance in three dosing experiments were 2.7-4.8 h, 37-64 mIikg, 59-98 mlJkg and 16-25 mUkglh respectively. Glycyrrhizin was not detected in plasma after oral administration of the usual therapeutic dose of glycyrrhizin and no dose dependency of the drug was observed in dose range of 40-120 mg (Yamamura et al., 1992). The effect of components of aqueous licorice root extract (LE) on the pharmacokinetics of glycyrrhizin and glycyrrhetic acid was investigated in rats and humans. The aim of the work was to define the role of pharmacokinetics on glycyrrhizin toxicity. Significantly lower glycyrrhizin and glycyrrhetic acid plasma levels were found in rats and humans treated with LE compared to the levels obtained with those in which glycyrrhizin alone was present. The pharmacokinetic curve showed significant differences in the area under the plasma-time curve (AUC), Cmax and Tmax parameters. The data obtained from urine samples were in agreement with the above results and confirm a reduced availability of glycyrrhizin present in LE compared to pure glycyrrhizin. This should be attributed to the interaction during intestinal absorption between the glycyrrhizin constituent and the several constituent of LE. The modified bioavailability could explain the
Pharmacokinetics and Drug Interactions of Glycyrrhizin I Glycyrrhiza
407
various clinical adverse effects resulting from the chronic administration of glycyrrhizin alone as opposed to LE (Cantelli-Forti et al., 1994) The pharmacokinetic behavior of glycyrrhizin in four patients with acute hepatitis (hepatitis group) and six patients with liver cirrhosis (cirrhosis group) receiving chronically an iv administration of a 120 mg dose once a day or once every other day of glycyrrhizin was investigated. The plasma concentration of glycyrrhizin declined monoexponentially in both groups. The elimination half life for glycyrrhizin in hepatitis and cirrhosis group varied significantly in the range of 2.7-7.6 h and 6.2-40.1 h and total body clearance (CL total) in the range of 2.8-23.2 mUh/kg and 1.4-12.9 mUh/kg respectively. The half life for glycyrrhizin in the hepatitis and cirrhosis group was about twice and eight times that in normal subjects respectively and CL total values were about 0.7 and 0.23 times that in normal subjects respectively. There was significant correlation between CLtotal and hepatic functions (serum aspartate aminotransferase and alanine aminotransferase) in both patient groups. With improvements of the liver function the CL total for glycyrrhizin increased from 2.8 mUh/kg to 11.4 mUh/kg and the half life shortened from 7.6 h to 3.4 h. These findings indicated that the variation of pharmacokinetic behavior of glycyrrhizin in both groups was closely related to the extent of the liver function (Yamamura et al., 1995). In a subsequent study pharmacokinetics of iv glycyrrhizin was evaluated after single and multiple doses in European patients with chronic hepatitis C infection. Patients received 80, 160 or 240 mg glycyrrhizin three times a week or 200 mg glycyrrhizin six times a week. Glycyrrhizin level was determined by HPLC. Decline in plasma glycyrrhizin concentration was monoexponentiaL Glycyrrhizin exhibited linear pharmacokinetics up to 200 mg dose. No significant differences in CL total and half life were observed between cirrhotic and non cirrhotic patients. No correlation between hepatic function and pharmacokinetics was found, probably because patients with severe liver disease were excluded in the study (Van Rossum et al., 1999). The intestinal bacteria, Eubacterium sp. and Bifidobacterium sp., participate in the metabolism of glycyrrhizin which is an active ingredient ofkampo-medicines. Most antibiotics have activity against intestinal bacteria. This means that antibiotics may lower the metabolism of glycyrrhizin when administered in combination. On the other hand, it is also highly possible that bacterial preparations such as Bifidobacterium longum, Clostridium butyricum and Streptococcus faecalis increase the number of Eubacterium sp. and Bifidobacterium sp., resulting in enhanced metabolism of glycyrrhizin when they are used concomitantly with kampo-medicines. The studies suggested that the drug interactions of kampo-medicines with antibiotics and bacterial preparations should be confirmed in clinical studies (Ishihara et al., 2002).
408
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
The influence of synthetic drugs prescribed for peptic ulcer on the pharmacokinetic fate of glycyrrhizin from Shaoyao-Gancao-tang (SGT, a traditional Chinese formulation) was investigated in rats. Co-administration of histamine H2-receptor antagonist (cimetidine) and.anticholinergic drug (scopolamine butyl bromide) with SGT didn't influence the area under the plasma concentration-time curves (AVC) of glycyrrhetic acid, an active metabolite derived from glycyrrhizin in SGT. The AVC of glycyrrhetic acid from SGT were significantly reduced by co-administration of synthetic drugs commonly used for peptic ulcer in a triple therapy (OAM), a combination of a proton pump inhibitor (omeprazole) and two antibiotics (amoxicillin and metronidazole). The reduction of AVC in OAM treatment was due to the antibacterial effect of amoxicillin and metronidazole on intestinal bacteria in rat which lead to the decrease of glycyrrhizin hydrolysis activity. The study suggests that it may not be a proper way to use triple therapy containing antibiotics simultaneously with SGT for healing of chronic ulcers (He et al., 2001). Another study indicates that, the bioavailability of glycyrrhizin in SGT is severely reduced by a single administration of amoxicillin and metronidazole (AMPC-MET), and the reducing effect continues for 12 days. In order to reduce the negative effect of AMPC-MET on the bioavailability of glycyrrhizin, the optimum scheduling of the medications was examined. It was found that the reduction in the plasma glycyrrhetic acid concentration and the glycyrrhizin -metabolizing activity in faeces caused by a single dose of AMPC-MET could be sharply attenuated by the repetitive administration of SGT for 4 days. The glycyrrhetic acid concentration and the glycyrrhizin-metabolizing activity were strongly enhanced by further continuous administration of SGT. These findings suggest that repetitive administration of SGT starting 1 or 2 days after the administration of AMPC-MET speeds the recovery of the bioavailability of glycyrrhizin in SGT. Such strategies for administering medications may also be useful for combination therapy of antibiotics with other traditional Chinese formulations containing bioactive glycosides (He et al., 2003). The pharmacokinetics of total and free prednisolone (PSL) in six healthy men with or without pretreatment with oral glycyrrhizin was investigated to confirm whether oral administration of glycyrrhizin influences the metabolism of PSL in man. Each subject received an iv administration of 0.096 mg/kg of prednisolone hemisuccinate (PSL-HS) with or without pretreatment with 50 mg oral glycyrrhizin four times. Total PSL and free PSL in plasma were measured by HPLC and isocolloid osmolar equilibrium dialysis method. The pharmacokinetic parameters of PSL were determined by non-compartmental analysis. Oral administration of glycyrrhizin was found to significantly increase the concentration of total PSL at 6,8 hand ofthe free PSL at 4,6 and 8 h after PSL-HS. After oral administration of glycyrrhizin the area under the curve (AVC) was
Pharmacokinetics and Drug Interactions of Glycyrrhizin / Glycyrrhiza
409
significantly increased, total plasma clearance (CLtotal) was significantly decreased and the mean residence time (MRT) was significantly prolonged. However the volume of distribution (Vdss) showed no evident change. This suggests that oral administration of glycyrrhizin increases the plasma PSLconcentrations and influences its pharmacokinetics by inhibiting its metabolism but not by affecting its distribution (Chen et al., 1991). Prolonged intake of licorice extract (LE) or glycyrrhizin on murine liver cyp catalyzed drug metabolism was studied. Results indicate that the induction of cytochrome P450 dependent activities by prolonged intake of high LE or glycyrrhizin doses may result in accelerated metabolism of coadministred drug with important implications for their disposition (Paolini et al., 1998). Pharmacokinetics of glycyrrhizin following oral and iv administration has been summarized below (Fig 2). Glycyrrhizin (p.o)
i Partly absorbed intact, from GIT (Minor)
i Not found in plasma
l
Metabolism by intestinal bacteria (Major)
~
l
Glycyrrhetic Acid
~
Found in urine
Absorbed
Found in plasma
Glycyrrhizin (i.v) (Exhibits biexponential profile during 24 h)
~
Transported to liver
~ Lysosomal beta 0 glucuronidase Glucuronide Conjugates Glycyrrhetic acid 3-0-beta-D-glucuronide (GAMG)
~
Transported in bile to intestine
~
Metabolism by intestinal bactena
Reabsorbed
Fig 2. Pharmacokinetics of glycyrrhizin following oral and iv administration
LICORICE DRUG INTERACTIONS Licorice has been found to interact with a number of drugs leading to effects varying from increased risk of bleeding, hypokalemia, elevated blood
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
410
pressure, increased risk of corticosteroid adverse effects and increased risk of digoxin toxicity. The following table (Table 1) gives an overview of the various dimensions of licorice interactions along with the probable mechanisms involved and clinical management of the adverse effects. Table 1. Licorice Drug Interactions Drugs
ALTEPLASE (for the treatment of acute ischemic stroke), ASPIRIN (NSAID), DIPYRIDAMOLE,( inhibits thrombus formation) PRA VAST ATIN SODIUM (HMGcoA reductase inhibitor), CLOPIDOGREL (potent oral anti platelet agent), DICUMAROL (Vitamin K antagonist). ANTACIDS, CASTOR OIL (strong laxative), PERU BALSAM, BISACODYL.
Effect of the interaction with licorice
Summary of the Management Mechanism interaction
The risk of Licorice may potentibleeding in- ate the effects of creases. anti platelet drugs (Norred & Brinker, 2001; Heck et al., 2000). Licorice root contains coumarins and derivatives of coumarin which are anticoagulants (Heck et al., 2000). Glycyrrhizin demonstrated selective thrombin inhibition activity in vitro (Francischetti et al., 1997).
Caution is primarily advised if licorice is to be taken along with an antiplatelet agent. The signs and symptoms of excessive bleeding should be monitored and dose of drug adjusted accordingly.
Hypokalemia may increase due to concurrent administration.
Concomitant Excessive pouse of licorice tassium loss. and laxatives should be avoided.
With concurrent use of licorice and laxatives hypokalemic paralysis has been reported (Hussain, 2003; Lin et al., 2003; Shintani et al., 1992; Corsi et al., 1983) Licorice alone could cause hypokalemia, hypertension, and cardiac arrhythmia. (Dellow et al., 1999; Eriksson et al., 1999; Kageyama et al. , 1997; Bernardi et al., 1994; Blachley & Knochel, 1980; Wash & Bernard, 1975).
Licorice causes inhibition of thrombin and platelet aggregation.
Table Contd.
Pharmacokinetics and Drug Interactions of Glycyrrhizin/Glycyrrhiza
411
Table 1. Contd. Drugs
ORAL CONTRACEPTIVE (Estrogen and progestogen combination).
Effect of the interaction with licorice Elevated blood pressure and increased risk of fluid retention.
Summary of the Management Mechanism
interaction
Elevated blood pressure and increased fluid retention is quite common with simultaneous use oflicorice and the oral contraceptives (Bernardi et al., 1994) which may be associated with the estrogen and/or progesterone. 3monoglucuronylglycyrrhetinic acid (3MGA), a metabolite of glycyrrhetinic acid inhibits ll-betahydroxysteroid dehydrogenase and reduces breakdown of cortisol, resulting in a hypermineralocorticoid effect (Kato et al., 1995; Walker & Edwards, 1994).
TESTOSTERONE Testosterone Endogenous test(steroid hormone). effectiveness osterone levels in healthy men and in decreases. women with polycystic ovary disease were significantly reduced when licorice was administered (Arm anini et al. , 1999; Takahashi & Kitao, 1994). It may inhibit conversion of androstenedione to testosterone through inhibition of 17-betahydroxysteroid dehydrogenase and 17, 20lyase (Arm anini et al., 1999; Sakamoto
If licorice is Mineralo-cor-
used with oral ticoid contraceptives effect and the pa- increased. tient develops fluid retention or hypertension, concurrent use should be discontinued.
Concomitant use of licorice and test osterone. should be avoided.
Theinhibition of 17-betahydr 0 xysteroid dehydr 0 gen ase and 17, 20lyase responsible for the catalytic conversion of androstenedione to testosterone; increase in the estradiol synthesis due to aromatase stimulation.
Table Contd.
412
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Table 1. Contd. Drugs
Effect of the interaction with licorice
BEl'AMRrHASONE, Chances CORTICOTROPIN, CORTISONE, DEXAMETHASONE.
of adverse effects of corticos t e r 0 i d Increase
Summary of the Management Mechanism interaction & Wakabayashi, 1988; Takeuchi et al., 1991). Some studies suggest that licorice may also stimulate aromatase activity and increase the estradiol to testosterone ratio. Glycyrrhizin increased the AUC of prednisolone and decreased the clearance in healthy subjects (Homma et al., 1994; Chen et al., 1990) The activity of topically applied hydrocortisone was potentiated by glycyrrhetic acid in healthy subjects (Teelucksingh et al., 1990).
Lower corticosteroid dose may be required to avoid adverse effects if licorice is to be used.
Corticosteroid metabolism inhibition by licorice.
Hypokalemia by caused licorice increase the risk of digoxin toxicity. Licorice ingestion causes pseudoald 0steronism resulting in hypokalemia.
DIGOXIN
Digoxin toxic- The symptoms of conity may gestive heart failure increase and hypokalemia appeared in a patient taking licorice, furosemide, and digoxin (Harada et al., 2002).
Concurrent use of licorice and digoxin must be avoided.
BENDROFLUMETHIAZIDE, BENZTHIAZIDE, BUMETANIDE, CHLOROTHIAZIDE, CHLORTHALIDONE, CYCLOTHIAZIDE (Potassium -losing diuretics).
Reduced effectiveness ofthe diuretic and increased risk of hypokalemia.
Concurrent use of licorice and potassium- losingdiuretics need to be avoided. Concurrent administration may need potassium supplement.
Some studies reported that patients experienced hypokalemia and hypertension with concomitant use of licorice and diuretics (Harada et al., 2002; Folkerson et al., 1996; Farese et al., 1991).
Pharmacokinetics and Drug Interactions of Glycyrrhizin I Glycyrrhiza
413
SUMMARY
Glycyrrhiza glabra (licorice) root contains a triterpenoid saponin called glycyrrhizin. Glycyrrhizin is being used in the treatment of hepatitis. Pharmacokinetics of glycyrrhizin has been evaluated in rats following intravenous iv and oral administration. Glycyrrhizin is a glycoside containing 2 glucuronic acid molecules attached to glycyrrhetic acid, the aglycone. Mter intravenous administration in rats, glycyrrhizin is secreted from liver into the bile and comes to intestine where it is hydrolyzed by intestinal bacteria to glycyrrhetic acid which is absorbed into the systemic circulation giving rise to a secondary peak in the elimination phase. Glycyrrhetic acid also is secreted from the liver as unchanged glycyrrhetic acid and glycyrrhetic acid monoglucoronide which is hydrolyzed in the intestine, reabsorbed and undergoes enterohepatic cycling thereby reducing clearance of the drug and prolonging its residence in the body. Thus decline in plasma concentration of glycyrrhizin, following iv administration is biexponential. Since secretion is an active process involving carriers, the transport of drug from liver to bile is a saturable process and pharmacokinetics of glycyrrhizin is nonlinear. Glycyrrhizin showed a dosedependent pharmacokinetics. Glycyrrhizin and glycyrrhetic acid both being actively secreted and having molecular weight >250, would have high biliary clearance. Oral absorption of glycyrrhizin is very poor (bioavailability 1%) probably due to its high molecular weight and polarity, it is slowly metabolized by the intestinal bacteria to glycyrrhetic acid and absorbed. Prolonged intestinal transit could increase the extent of absorption of glycyrrhetic acid from the gut. Intraperitoneal and nasal administration in rats has been found to improve bioavailability of glycyrrhizin. Studies in human subjects indicate oral glycyrrhizin is partly absorbed from gastrointestinal tract in intact form; rest of the dose is metabolized to glycyrrhetic acid and absorbed. The decline in plasma concentration following iv administration of glycyrrhizin is biexponential and no dose dependency of drug disposition has been observed. Glycyrrhizin iv administration in cirrhosis/hepatitis patients showed a monoexponential decline of drug in plasma indicating a linear pharmacokinetics of the drug and the pharmacokinetics are closely related to the extent of liver function. Similarly pharmacokinetics of glycyrrhizin iv, in patients with chronic hepatitis C infection, has also been linear but no correlation between hepatic function and pharmacokinetics is observed which may be explained by the patients having milder liver disease. Oral administration of amoxicillin and metronidazole could destroy intestinal bacteria and thereby make co administered glycyrrhizin ineffective by inhibiting its hydrolysis to glycyrrhetic acid. Glycyrrhizin could inhibit hepatic metabolism of corticosteroids like prednisolone resulting in toxicity. Licorice (glycyrrhizin) if concurrently administered with anticoagulant could cause bleeding due to its antiplateletlantithrombin effect. Concurrent administration oflicorice
414
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
with potassium-losing diuretics, laxatives cause excessive potassium loss and may need potassium supplement. Licorice induced hypokalemia may cause digoxin toxicity. Licorice in combination with contraceptive may cause sodium and fluid retention and elevated blood pressure. In human subjects, oral absorption of glycyrrhizin from aqueous licorice root extract has been found to be lower than that obtained after administration of pure glycyrrhizin. Thus prolonged administration of pure glycyrrhizin carries more chances of adverse effects as opposed to crude licorice root extract.
REFERENCES Akao, T., Hayashi, T., Kobashi, K., Kanaoka, M., Kato, H., Kobayashi, M., Takeda, S. and Oyama, T. (1994). Intestinal bacterial hydrolysis is indispensable to absorption of 18 beta-glycyrrhetic acid after oral administration of glycyrrhizin in rats. J Pharm Pharmacol., 46(2): 135-137. Armanini, D., Bonanni, G. and Palermo, M. (1999). Reduction of serum testosterone in men by licorice. N Engl J Med., 341(15): 1158. Bernardi, M., D'Intino, P.E., Trevisani, F. et al. (1994). Effects of prolonged ingestion of graded doses of licorice by healthy volunteers. Life Sci., 55(11): 863-872. Blachley, J.D. and Knochel, J.P. (1980). Tobacco chewer's hypokalemia: licorice revisited. N. Engl. J. Med., 302(14): 784-785. Cantelli-Forti, G., Maffei, F., Hrelia, P., Bugamelli, F., Bernardi, M., D'Intino, P., Maranesi, M. and Raggi, M.A. (1994). Interaction of licorice on glycyrrhizin pharmacokinetics. Environ Health Perspect., 102(9): 65-68. Chen, M.F., Shimada, F., Kato, H. et al. (1990). Effect of glycyrrhizin on the pharmacokinetics of prednisolone following low dosage of prednisolone hemisuccinate. Endocnnol Jpn., 37: 331-341. Chen, M.F., Shimada, F., Kato, H., Yano, S. and Kanaoka, M. (1991). Effect of oral administration of glycyrrhizin on the pharmacokinetics of prednisolone. Endocrinol Jpn., 38(2): 167-174. Corsi, F.M., Galgani, S., Gasparini, C. et al. (1983). Acute hypokalemic myopathy due to chronic licorice ingestion: report of a case. Ital J Neurol Sci., 4(4): 493-497. Dellow, E.L., Unwin, RJ. & Honour, J.W. (1999). Pontefract cakes can be bad for you: refractory hypertension and liquorice excess. Nephrol Dial Transplant., 14: 218-220. Eriksson, J.W., Carlberg, B. and Hillorn, V. (1999). Life-threatening ventricular tachycardia due to liquorice-induced. J Int Med., 245(3): 307-310. Farese, RV., Biglieri, E.G. and Shackleton, C.H.L. et al. (1991). Licorice-induced hypermineralocorticoidism. N Eng J Med., 325(17): 1223-1227. Folkerson, L., Knudsen, N.A. and Teglbjaerg, P.S. (1996). Licorice. A basis for precautions one more time! Ugeskr Laeger, 158(51): 7420-7421. Francischetti, LM., Monteiro, R.Q. and Guimaraes, J.A. (1997). Identification of glycyrrhizin as a thrombin inhibitor. Biochem Biophys Res Commun., 235(1): 259-263. Harada, T., Ohtaki, E., Misu, K., et al. (2002). Congestive heart failure caused by digitalis toxicity in an elderly man taking a licorice-containing Chinese herbal laxative. Cardiology, 98(4): 218. He, J,X" Akao, T., Nishino, T. and Tani, T. (2001). The influence of commonly prescribed synthetic drugs for peptic ulcer on the pharmacokinetic fate of glycyrrhizin from ShaoyaoGancao-tang. Biol Pharm Bull., 24(12): 1395-1399. He, J.X., Akao, T. and Tani, T. (2003). Repetitive administration of Shaoyao-Gancao-tang to rats restores the bioavailability of glycyrrhizin reduced by antibiotic treatment. J Pharm Pharmacol., 55(11): 1569-1575. Heck, Amy, M., Beth, A., Lukes and Anita, L. (2000). Potential interactions between alternative therapies and warfarin. American Journal ofHealth-System Pharmacy, 57(13): 1221-1230.
Pharmacokinetics and Drug Interactions of Glycyrrhizin / Glycyrrhiza
415
Homma, M., Oka, K., Niitsuma, T., et al. (1994). A novel 11-beta-hydroxysteroid dehydrogenase inhibitor contained in Saiboku-To, a herbal remedy for steroid-dependent bronchial asthma. J Pharm Pharmacal., 46(4): 305-309. Hussain, R.M. (2003). The sweet cake that reaches parts other cakes can'tl Postgrad Med J. 79: 115-116. Ichikawa, T., Ishida, S., Sakiya, Y., Sawada, Y. and Hanano, M. (1986). Biliary excretion and enterohepatic cycling of glycyrrhizin in rats. J Pharm Sci., 75(7): 672-675. Ishida, S., Sakiya, Y., Ichikawa, T. and Awazu, S. (1989). Pharmacokinetics of glycyrrhetic acid, a major metabolite of glycyrrhizin, in rats. Chem Pharm Bull. (Tokyo), 37(9): 2509-2513. Ishida, S., Sakiya, Y., Ichikawa, T. and Taira, Z. (1992). Dose-dependent pharmacokinetics of glycyrrhizin in rats. Chem Pharm Bull. (Tokyo), 40(7): 1917-1920. Ishihara, M., Honma, M., Kuno, E., Watanabe, M. and Kohda, Y. (2002). Combination use of kampo-medicines and drugs affecting intestinal bacterial flora. Yakugaku Zasshi., 122(9): 695-701. Kageyama, K., Watanobe, H., Nishie, M., et al. (1997). A case of pseudo aldosteronism induced by a mouth refresher containing licorice. Endocr J., 44(4): 631-632. Kato, H., Kanaoka, M., Yano, S. et al. (1995). 3-Monoglucuronyl-glycyrrhetinic acid is a major metabolite that causes licorice-induced pseudoaldosteronism. J Clin Endocrinol Metab., 80(6): 1929-1933. Kawakami, J., Yamamura, Y., Santa, T., Kotaki, H., Uchino, K., Sawada, Y. and Iga, T. (1993). Kinetic analysis of glycyrrhetic acid, an active metabolite of glycyrrhizin, in rats: role of enterohepatic circulation. J Pharm Sci., 82(3): 301-305. Lin, S.H., Yang, S.S., Chau, T. et al. (2003). An unusual cause of hypokalemic paralysis: chronic licorice ingestion. Am J Med Sci., 325(3): 153-156. Merck, Index (2006). 14th Edition, Merck & Co. INC., NJ, USA. Norred, C.L. and Brinker, F. (2001). Potential coagulation effects of preoperative complementary and alternative medicines. Altern Ther Health Med., 7(6): 58-67. Paolini, M., Pozzetti, L., Sapone, A. and Cantelli-Forti, G. (1998), Effect of licorice and glycyrrhizin on murine liver CYP-dependent monooxygenases. Life Sci., 62(6): 571-582. Reynolds, J.E.F. (1989). Martindale: The Extra Pharmacopoeia. 29 th ed. London: The Pharmaceutical Press. Sakamoto, K. and Wakabayashi, K. (1988). Inhibitory effect of glycyrrhetinic acid on testosterone production in rat gonads. Endocrinol Jpn., 35: 333-342. Sasaki, K., Yonebayashi, S., Yoshida, M., Shimizu, K., Aotsuka, T. and Takayama, K. (2003). Improvement in the bioavailability of poorly absorbed glycyrrhizin via various non-vascular administration routes in rats. Int J Pharm., 265(1-2): 95-102. Shintani, S., Murase, H., Tsukagoshi, H. et al. (1992). Glycyrrhizin (Licorice)-induced hypokalemic myopathy. Eur Neural., 32(1): 44-51. Takahashi, K. and Kitao, M. (1994). Effect of TJ-68 (Shakuyaku-Kanzo-To) on polycystic ovarian disease. Int J Fertil., 39(2): 69-76. Takeda, S., Ishthara, K., Wakui, Y., Amagaya, S., Maruno, M., Akao, T. and Kobashi, K. (1996). Bioavailability study of glycyrrhetic acid after oral administration of glycyrrhizin in rats; relevance to the intestinal bacterial hydrolysis. J Pharm Pharmacol., 48(9): 902-905. Takeuchi, T., Nishii, 0., Okamura, T., et al. (1991). Effect of paeoniflorin, glycyrrhizin and glycyrrhetic acid on ovarian androgen production. Am J Chin Med., 19(1): 73-78. Teelucksingh, S., Mackie, A.D., Burt, D. et al. (1990). Potentiation of hydrocortisone activity in skin by glycyrrhetinic acid. Lancet, 8697: 1060-1063. Tsai, T.H., Liao, J.F., Shum, A.Y. and Chen, C.F. (1992). Pharmacokinetics of glycyrrhizin after intravenous administratiton in rats. J Pharm Sci., 81(9): 961-963. van Rossum, T.G., Vulto, A.G., Hop, W.C. and Schalm, S.W. (1999), Pharmacokinetics of intravenous glycyrrhizin after single and multiple doses in patients with chronic hepatitis C infection. Clin Ther., 21(12): 2080-2090. Vyas, Manju and Majumdar, D.K. (2005). Glycyrrhizin- A Bioactive Principle from Glycyrrhiza glabra. In: Recent Progress in Medicinal Plants, Vol. 9, Ed. By Majumdar,
416
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
D.K., Govil, J.N., Singh, V.K. and Sharma, R.K., Studium Press LLC, USA, pp. 119136. Walker, B.R. and Edwards, C.R.W. (1994). Licorice-induced hypertension and syndromes of apparent mineralocorticoid excess. Endocrinol Metab Clin N Am., 23: 359-377. Wash, L.K. and Bernard, J.D. (1975). Licorice-induced pseudoaldosteronism. Am J Hasp Pharm., 32(1): 73-74. Yamamura, Y., Kawakami, J., Santa, T., Kotaki, H., Uchino, K., Sawada, Y., Tanaka, N. and Iga, T. (1992). Pharmacokinetic profile of glycyrrhizin in healthy volunteers by a new high-performance liquid chromatographic method. J Pharm Sci., 81(10): 1042-1046. Yamamura, Y., Santa, T., Kotaki, H., Uchino, K., Sawada, Y. and Iga, T. (1995). Administration-route dependency of absorption of glycyrrhizin in rats: intraperitoneal administration dramatically enhanced bioavailability. BioI Pharm Bull., 18(2): 337-34l. Yamamura, Y., Tanaka, N., Santa, T., Kotaki, H., Aikawa, T., Uchino, K., Osuga, T., Sawada, Y. and Iga, T. (1995). The relationship between pharmacokinetic behaviour of glycyrrhizin and hepatic function in patients with acute hepatitis and liver cirrhosis. Biopharm Drug Dispos., 16(1): 13-21.
21 Determination of Total Antioxidant Status and Rosmarinic Acid from Orthosiphon stamineus in Rat Plasma GABRIEL AKYIREM AKOWUAH 1,* AND ISMAIL ZHARI2
ABSTRACT A simple high-performance liquid chromatography (HPLC) method was developed to determine the content of rosmarinic acid (RA) in plasma of rat collected at different time (0-8 h) after oral administration of Orthosiphon stamineus leaf extract. The features of the assay include protein precipitation using acetonitrile, isocratic elution and reverse phase C-18 column with ultraviolet (UV) detection. The maximum concentration of RA in the plasma was 1.10 ± 0.34 JlglmL at 1 h after oral administration of the extract. The extract showed significant reduction (p<0.05) of malondialdehyde (MDA) levels in plasma samples of rats fed with the methanol extract compared with a control group. The total antioxidant status in plasma was significantly (p<0.05) elevated in rats treated with the methanol extract compared to normal controls. Key words : Antioxidant status, lipid peroxidation, Orthosiphon stamineus, plasma, rosmarinic acid
INTRODUCTION Rosmarinic acid (RA) is an ester of caffeic acid and 3,4dihydroxyphenyllactic acid, which are important natural bioactive substances occurring widely in food plants (Ro et al., 1992). It is a well 1. School of Pharmacy, University College Sedaya International, 56000, Kuala
Lumpur, Malaysia. 2. School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia. * Corresponding author : E-mail: [email protected]
418
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
known natural product extracted from rosemary plant (Rosmarinus officinalis), and other members of Labiatae (Litvinenko et al., 2001). It occurs throughout the Boraginaceae, Lamiaceae, Zosteraceae (Takeda et al., 1990) and in lower plants such as the hornworts (Ravn et al., 1994). RA has a various medicinal values and well characterized physiological functions. The main activities are astringent, antioxidative, antiinflammatory, antimutagen, antibacterial and antiviral (Parnham & Kesselring, 1985). RA obtained from plants is a multi-active substance used in cosmetics to maintain healthy skin due to its antioxidant qualities which is superior to that of vitamin E (Leung & Foster, 1996). Generally, after administration of a drug, large amount distributes throughout the body and resided in the body at various times, some leaving soon after absorption whilst others taking a longer time to eliminate. Polyphenolic substances such as phenolic acids and flavonoids are reported to be degraded by gut flora and then metabolized in tissues such as kidneys, liver and intestine (Williamson et al., 2000; Rechner et al., 2002). Orthosiphon stamineus has several chemically active constituents including RA, the most abundant phenolic acid in the leaves. There is scarcely any report on the absorption studies of bioactive polyphenolic acid constituents in O. stamineus leaf due to the complexity of these components in the leaf hence this report describes a simple, rapid and validated HPLC method for the determination of RA from O. stamineus leaf extract in rat plasma after oral administration. The study also includes evaluation of plasma lipid peroxidation inhibition and total plasma antioxidant status in rats.
MATERIALS AND METHODS Chemical and Reagents Rosmarinic acid was purchased from Sigma Chemical Company (St. Louis MO, USA). 2,2'-azino-di[3-ethylbezthiazoline sulfonate (ABTS), thiobarbituric acid (TBA), trichlroacetic acid, 1,1,3,3-tetraethoxypropane were obtained from Sigma (USA). Methanol, acetonitrile, and water (acidified to pH 3 with phosphoric acid), were obtained from Merck (Darmstadt, Germany). Membrane filters (0.45 lIm pore size) from Millipore were used for filtration of the mobile phase and the samples. All solvents were of analytical grade or HPLC grade.
Plant Samples Plants were grown from cuttings using standard agronomic practices at Kepala Batas (Pinang, Malaysia). The cultivated leaves were collected in late afternoon, from 30- to 45-day-old plants. Voucher specimen of the plant material was deposited at Bilik Herba, School of Pharmaceutical Sciences, Universiti Sains Malaysia.
Antioxidant Status and Rosmarinic Acid from Orthosiphon stamineus
419
Preparation of Extracts Powdered dried leaves ofA. paniculata (1.0 kg) were extracted with methanol using soxhlet. The extract was filtered (Whatman No.1) with a Buchner filter and concentrated under vacuum. 100 g of the methanol extract was freeze-dried. The content of RA in the freeze dried extract was determined by HPLC before oral administration.
Animals and Dosage Experimental animals used were Sprague-Dawley rats, weighing 220-250 g, from the Animal House of University Sains Malaysia. The animals were kept in the animal room ( 25 ± 2 DC) under 12 h light/dark cycle and fed with standard diet and free access to distilled water prior to the start of the study. Animals were maintained and handled according to the recommendations of the local ethical committee which approved the design of the animal experiments.
Sampling of Blood Six overnight fasted rats were treated orally with single dose of 0. stamineus freeze-dried extract at 1 g/kg body weight in 10 mL ofthe extract suspension. The freeze-dried extract was dissolved in distilled water and vortex prior to oral administration. The animals were put under anaesthesia with sodium pentobarbital (50 mg/kg i.p.) and remained anesthetized throughout the experimental period. The jugular vein was cannulated with Pe-50 tubing to collect blood with an established heparin-lock using 100 U/mL heparin in saline. Blood samples (0.3 mL) were taken using a disposable syringe at o min (pre-dose) 0.5, 1, 2, 3, 4, 6 and 8 h post dose. Blood samples were immediately transferred to a heparinized microcentrifuge tube and centrifuged at 3000 rpm for 10 min at 5°C. The resulting plasma sample (0.2 mL supernatant) was transferred into 1.5 mL tubes and stored at -80 DC until assayed.
Preparation of Standard Solutions of Plasma The stock solution 0.1 mglmL of the reference RA was freshly prepared in acetonitrile. Solutions ofRA in the range of 0.1- 10 !J,glmL were prepared in blank plasma samples for calibration curve. Spiked plasma samples were vortexed for 15 seconds and centrifuged at 3000 x g for 5 min. at 5°C. The resulting supernatants were used for injection. Quality control (QC) samples at the concentration of 0.1 J.lglmL, 1 J.lglmL and 10 J.lglmL were prepared by the same procedures as described.
HPLC Conditions for RA in Plasma The plasma (0.2 mL) was precipitated with acetonitrile (0.3 mL) and centrifuged at 3000 x g for 10 min. at 5°C. The organic layer was transferred into an empty tube and dried under a stream of nitrogen at 40°C. The
420
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
residue was reconstituted in acetonitrile to give a 0.5 mL solution. The solution was passed through membrane filter (pore size 0.5 pm,) prior to HPLC analysis. HPLC analysis was performed with a Gilson HPLC pump (Model 305), a Gilson UVNIS detector (Gilson Medical Electronics, VillierssIe-Bel, France) connected to a Hitachi D-2500 Chromato-integrator (Hitachi, Tokyo, Japan) and a Rheodyne sample injector valve fitted with a 50 pI sample loop. The detector was operated using a sensitivity range of 0.005 AUFS. A LiChrosorb RP-18 column (250 mm x 4.6 i.d. mm, 10 pm particle size) (Merck) was used. The chromatographic conditions used for analyzing plasma samples were; Flow-rate of 1 mUmin; Injection volume of 20 Ill; UV detection at 340 nm and ambient temperature for all HPLC analysis. The mobile phase for elution ofRA was methanol: water (pH = 3) (8:2; v/v).
Validation of HPLC Method
Linearity and sensitivity: The linearity of the responses was determined for seven concentrations in plasma by three injections. The contents of RA were calculated using regression parameters obtained from the standard curve for blank plasma samples. The limit of quantification (LOQ) was established at signal to noise ratio (SIN) of 10. Precision Intra- and inter-day precision of the assay were determined five times on the same day and continuously for 5 days at the concentration of the QC solutions. The intra- and inter-assay relative standard deviation was used to validate the precision of the assay by determining standard samples of RA in plasma.
Accuracy Accuracy was determined by recovery studies. Recoveries ofRA from plasma were estimated by comparing the area obtained from injections of standard solutions (QC solutions) in blank plasma to those in acetonitrile. The mean recoveries were determined at the concentration of the QC solutions in triplicates.
Stability The stability of the reference compound was studied in the plasma and acetonitrile precipitated plasma with the QC samples. The samples were put aside at room temperature for 12 h, or stored in a refrigerator at 4°C and -20°C for 1, 2, and 3 days; -80°C for 1 month. The content of RA was calculated. The compound was considered stable if the variation of the content was less than 10% of initial time concentration or response (Hu & Morris, 2003).
Antioxidant Status and Rosmarinic Acid from Orthosiphon stamineus
421
Data Analysis
Calibration Curves Standard calibration curve for RA was prepared and the weight was computed with equation obtained from linear regression analysis; y=mx+b where y = relative peak area of analyte, m = slope of the line generated by a standard curve; x = concentration of analyte found (~mL); b = intercept of the line generated by the standard curve. Amount of RA in plasma: The amount of analyte found in plasma (ng/mL) was calculated as follows: A = C x Vfl Vp where A =ng/mL of the analyte found in test sample; C = concentration in Ilg/~L of the analyte found in test samples from standard curve; Vp = test portlOn (0.2 mL, plasma); volume Vf = final volume ( 0.5 mL, plasma).
Pharmacokinetics Parameters The following pharmacokinetic parameters were determined for the period of 0-8 h: the area under plasma concentration time curve from time zero to the last measurable RA sample time (AUC O_8 h); maximum plasma concentration (C max ) and the time to reach C max (Tmax) were determined directly from the data. The elimination rate constant Kel (h- 1 ) was calculated by log /linear regression using terminal phase of the plasma concentration time plot. The half-life, t 1J2 , (h) was calculated by dividing 0.693 by Kel.
Lipid Peroxidation (MDA assay) and Plasma Antioxidant Status Experimental animals used were Sprague-Dawley rats, weighing 220-250 g. The rats were separated into 2 groups with each group containing six animals (n = 6). The animals in the first group were treated orally with single dose of 1 g/kg body weight in 10 mL of the extract suspension per day for two weeks. The animals in the second group served as a control were given appropriate amount of distilled water. 24 h after the administration of the last dose of the extract, the blood samples were collected from the orbital plexus. Blood samples were immediately transferred to a heparinized micro centrifuge tube and centrifuged at 3000 rpm for 10 min. at 5°C. The resulting plasma sample (supernatant) stored at -80°C until assayed. Rats were not deprived of feed before obtaining their blood samples to prevent possible changes in plasma antioxidant status cause by latent stresses of food deprivation. Thiobarbituric acid reactive substances (TBARS) content of the plasma samples was determined by the method of Drapper et al. (1993). The amount of TBARS produced was calculated using 1,1,3,3, tetraethoxypropane as a standard. Lipid peroxidation was expressed in nmoles ofTBARS formed per mL ofthe sample. Plasma antioxidant status of the samples was determined
422
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
by the method of described by Miller et al. (1993). Trolox was used as a standard. The result was expressed as nmoles of trolox equivalent antioxidant capacity (TEAC) per litre of sample. Data are presented as means ± S.D. (n=6). Student's t-test was used for statistical significance between groups. P values<0.05 were considered statistically significant.
RESULTS HPLC Assay Development and Validation The RA (Fig 1) content of the freeze dried extract determined prior to oral administration to the animals was 4.10% w/w. External standard HPLC method was used to analyze RA in plasma samples following the oral administration of o. stamineus (1 g/kg body weight) to rat. As shown in Fig 2, RA (t R =14.8 min) in the plasma was well resolved by using a mixture of methanol: water (pH = 3) (8:2). Deproteinization of plasma samples with acetonitrile resulted in less interference from endogenous compounds, best accuracy, precision and recovery. The HPLC method was validated; parameters included in the method validation were linearity, accuracy, precision and limit of quantification. The calibration curves were linear over the concentration range of 0.1-10 J.lg/mL with correlation coefficient greater than 0.998 in all standard curves. The limit of quantification (LOQ) of RA determined in blank rat plasma samples at signal to noise ratio of 10 was 50 ng/mL. The intra- and inter-assay relative standard deviation was used to validate the precision of the assay. Table 1 shows the results of inter- and intra-day precision and accuracy for RA in rat plasma. Acceptable precision was achieved with the method as revealed by the R.S.D. data, which did not exceed 10%. The mean recoveries determined at the QCs in triplicates were between 90.64 - 95.12%. The R.S.D. of recovery from plasma was less than 10%. The compound was stable at room temperature, -20°C and 4°C in plasma. No peaks corresponding to degradation products were observed.
~ OyOII(c'-OH
HO
~ : : : :,. .
I
O~
OH
Fig 1. Structure of rosmarinic acid
Antioxidant Status and Rosmarinic Acid from Orthosiphon stamineus
423
.. N
"$
•• •• ••
RA
\
•• •• ••
\
\
RA
\
." ." ...., .., ...
RA
..
111 IL IL
VI
o
L L
o
"
...... II>
o
~ ~
.•
lI-
n
." 0:
w 11111111111111111111
1'1
•
I U
fl
N
.,
III
:t
n
lSI
1111 111111 11111 11111
II:
II
U
1'1
N
u
a
~
11111111111111111111
$
I
U
$
J)
N
1111111111111111111111
..,
~
n
s
N
c·
c
b
5
l:
W
d
Fig 2. Chromatograms for rat plasma samples obtained from the analyses of rosmarinic acid (RA) from 0. stamineus, lal blank plasma, [bl blank plasma spiked with RA, [cl a rat plasma sample obtained after an oral dose of 19/kg of the extract. (d) Methanol leaf extract. Chromatographic peaks were identified with the aid of pure reference standard based on retention time tR Table 1. Precision and accuracy of rosmarinic acid in rat plasma Precision (n=5) Concentration added (}lglmL) 0.1 1
10
Accuracy (n=3)
Intra-day
Inter·day
RecoveryB
(RSD,%)
(RSD,%)
(%)
RSDb (%)
5.12 4.39 2.73
5.60 3.85 2.47
90.64 92.25 95.12
9.65 8.74 6.89
RSD: relative standard deviation in percentage Recovery = (calculated condspike conc) x 100. b RSD = (SD/conc ) x 100 a
The validated method was used for determination of RA in rat plasma samples after oral administration of O. stamineus extracts. The time course of plasma concentrations of RA after oral administration of the extract is shown in Fig 3. The parameters estimated from the curve included, t1l2 = 2.28 ± 0.30 h, Tmax = 1 h, C max = 1.10 ± 0.35 llg/mL and AUC O_8 h = 4.87 ± 1.56 Jlg/mL h.
424
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
~
1200
.@
!
.I 900 J I
o
o
2
4 6 Plasma collection time (h)
8
Fig 3. Plasma concentration-time curve of rosmarinic acid after oral administration of Orthosiphon stamineus with a single dose of 1 g/kg in rats. Data are means ± S.E. (n=6)
Lipid Peroxidation (MDA assay) and Total Antioxidant Status Fig 4 shows the effect of 14 days treatment of rats with single dose of the extract (1 g/kg of body weight) per day on malondialdehyde (MDA) levels and antioxidant status in plasma. The MDA levels were significantly reduced (p<0.05) in plasma samples of rats fed with the methanol extract compared with the control group. Significant lipid peroxidation inhibition of 30.2% as compared to the control group was observed in rats treated orally with the extract. The total antioxidant status in plasma was significantly 2
o
l---=~L-~_
Control
Extract
Fig 4. Effect of methanolic extract of Orthosiphon stamineus on plasma MDA levels (A) and plasma total antioxidant status (B) of rats treated orally with single .. dose of 1 g/kg body weight in 10 mL of the extract suspension for two weeks. Data are means ± S.D. (n=6). Student's t-test was used for statistical significance between groups. * p<0.05 as compared to control group
Antioxidant Status and Rosmarinic Acid from Orthosiphon stamineus
425
elevated in rats treated with the methanol extract compared to normal controls. An average increase of 11.3% in total antioxidant status was observed in plasma of rats treated orally with the extract.
DISCUSSION In our earlier studies, the methanolic extract of O. stamineus leaves was reported to exhibit significant radical scavenging activity in many in vitro models (Akowuah et al., 2005). In the present study, the methanol extracts showed lipid peroxidation inhibition and plasma antioxidant effect which may be ascribed to its polyphenol content. The aqueous methanolic extract of O. stamineus leaf has been reported to contain 67% of total identified phenolics (Sumaryono et al., 1991). Polyphenols have been observed to be very important in prevention of tissue damage by activated oxygen species due to their antioxidant effects including reactive oxygen species scavenging, metal chelation and enzyme modulation (Pietta et al., 1998). Naturally occurring polyphenols were shown to be effective in reducing oxidative stress (Holman & Katan, 1999). Rosmarinic acid, the most abundant polyphenolic acid in O. stamineus leaf extract (Akowuah et al., 2004), was reported to show antioxidant qualities superior to that of vitamin E and butylated hydroxytoluene (Leung & Foster, 1996). The increase in total antioxidant status and decrease in lipid peroxidation of rat plasma after 14 days treatment with extract of O. stamineus suggests that active constituents in the extract were absorbed. Absorption ofRA from the O. stamineus leaf extract was studied in order to gain more understanding on the oral administration ofthe extract in relation to the in vivo antioxidant effect. The bioavailability of RA is due to its hydrophilic character which facilitated its absorption and rapid eliminated from the blood circulation after intravenous administration (Parnham & Kesselring, 1985). RA and other polyphenolics have the tendency to bind to proteins and polysaccharide by hydrogen bonding because of the overall strength derived from multiplicity of hydrogen bonds radiating from the polyphenolic substrate. That is, absorption ofRA depends on factors related to the basic structure and since it is susceptible to degradation by gut microorganisms, the enzymes in body tissues after absorption, and other metabolic transformations distribution in the liver, bile, lung etc, only small proportion of RA reached the blood stream (Bravo et al., 1994; Baba et al., 2005). The result obtain in the present study agrees with these reports. The values of the pharmacokinetic parameters observed in this study may indicate absorption of some amount of free RA into the blood stream and hence distribution into tissues. The absorption of free RA into the b~ood circulation may account for the effective in vivo activity ofthe O. stamineus extract observed in the study. That is, some of the antioxidant activity of free RA remained during the metabolic process and may have affected the
426
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
plasma lipid peroxidation inhibition and antioxidant status as shown by our in vivo data. Metabolic transformations of drugs are believed to produce significantly less active metabolites compared to the parent drug. However, modifications of polyphenols by enterohepatic circulation mechanism do produce metabolites with considerable antioxidant properties. For instance, RA has been reported to be present as free, degraded and conjugated forms such as m-hydroxyphenylpropionic acid, m-coumaric acid, and sulfated form of caffeic acid and ferulic acid after oral administration of RA in rats and humans (Nakazawa & Ohasawa, 1988; Baba Osakabe et al., 2004). These metabolites are important natural antioxidant substances present in food plants hence they may have contributed substantially to the m vwo antioxidant activity.
CONCLUSIONS Antioxidant activity of herbals depends on the absorption of specific active constituents; therefore understanding of the in vivo antioxidant activity of O. stamineus leaf extract was attained by studying the absorption of the active ingredient (RA) in the extract after oral administration to rats. Most herbal medicines are administered orally in the form of crude extract in clinical use hence the absorption of RA in the gastrointestinal tract could be used as suitable reference in clinical application of herbal medicinal products from O. stamineus.
ACKNOWLEDGEMENTS The study was supported by Intensifying Research Prioty Areas (lRPA) Grant from Ministry of Science and Technology, and Environment, Malaysia. We acknowledge Dr. Nashiru Billa, School of Pharmacy, University of Nottingham, Malaysia Campus, for his support on the pharmacokinetic study.
REFERENCES Akowuah, A.G., Zhari, I., Norhayati, I. and Sadikun, A. (2004). Sinensitin, eupatorin, 3'hydroxy-5, 6, 7, 4'-tetramethoxyflavone and rosmarinic acid contents and antioxidative effect of Orthosiphon stammeus from Malaysia. Food Chemistry, 87: 569-566. Akowuah, A.G., Zhari, 1., Norhayati, 1. and Sadikun, A. (2005). Radical scavenging activity of methanolic extracts of Orthoslphon stamineus. Pharmaceutical Biology, 42: 629-635. Baba, S., Osakabe, N., Natsume, M. and Terao, J. (2004). Orally administered rosmarinic acid is present as conjugated and/or methylated forms in plasma, and is degraded and metabolized to conjugated forms of caffeic acid, ferulic acid and m-coumaric acid. Life Science, 75: 165-178. Baba, S., Osakabe, N., Natsume, M., Yasuda, A., Muto, Y., Riyoshi, K., Takano, R., Yoshikawa, T. and Terao, J. (2005), Absorption metabolism, degradation and urinary excretion of rosmarinic acid after intake of Perilla frustescens extract in humans. European Journal of Nutntwn, 44: 1-9.
Antwxidant Status and Rosmarinic Acid from Orthosiphon stamineus
427
Bravo, L., Abia, R., Eastwood, M.A. and Saura-Calixto, F. (1994). Nutritional antioxidants: Mechanisms, action, analyses of activities and medical applications. BrLtam Journal of NutrLtion, 71: 933-946. Draper, H.H., Squires, E.J., Mahmoodi, H., Wu, J., Agarwal, S. and Hardley, M. (1993). A comparative evaluation of thiobarbituric acid methods for the determination of malondialdehyde in biological materials. Free Radical Biology Medlcme, 15: 353-363. Ho, C.T., Lee, C.Y. and Huang, M.T. (1992), Phenolic compounds in Food and Their Effects on Health (pp. 506-507). Washington D.C.: ACS Symposium Series, ACS Press. Holman, P.C. and Katan, M.B. (1999), Dietary flavonoids; intake, health effects and bioavailability. Food Chemlcal Toxicology, 37: 937-942. Hu, H.E. and Morris, M.E. (2003). Determination of alpha-naphthyisothiocyanate in rat plasma and urine by high-performance liquid chromatography. Journal of Chromatography B., 788: 17-28. Leung, AY. and Foster, P. (1996). Encyclopaedw of Common Natural Ingredients Used in Foods, Drugs and Cosmetics, (pp. 446-448). New York: John Wiley and Sons. Litvinenko, V.I., Popova, T.P., Simonjan, AV., Zoz, I.G. and Sokolov, V.S. (1975). "Gerbstoffe' und OxyzimtsaureabkommLinge in Labiaten. Planta Medlca, 27: 372-380. Miller, N.J., Rice-Evans, C.A, Davies, M.J., Gopinathan, V. and Milner, A (1993). A novel method for measuring antioxidant capacity and its application to monitoring the antioxidant status in premature neonates. Clinical Science, 84: 407-412. Nakazawa, T. and Ohasawa, K. (1988). Metabolism of rosmarinic acid in rats. Journal of Natural Product, 61: 993-996. Parnham, J. and Kesselring, M. (1985), Rosmarinic acid. Drugs of the Future, 10: 756-757. Pietta, P., Simonetti, P., Gardana, C., Brusamolino, A., Morazzoni, P. and Bombardeli, E. (1998). Relationship between rate and extent of catechin absorption and plasma antioxidant status. Biochemical Molecular Bwlogy Internatwnal, 46: 895-903. Ravn, H., Pedersen, M.F., Andary, J., Borum, C., Anthoni, D., Christophersen, C. and Nielsen, P.H. (1994). Seasonal variation and distribution of two phenolic compounds, rosmarinic acid and caffeic acid, in leaves and roots-rhizomes of eegrass (Zostera marina 1.). Ophelw, 40: 51-61. Rechner AR., Kuhnle, G., Bremner P., Hubbard, G.P., Moore, K.P. and Rice-Evans, C.A (2002). The metabolic fate of dietary polyphenols in humans. Free Radical Biology Medicine, 33: 220-235. Sumaryono, W., Proksch, P., Wray, V., Witte, L. and Hartmann, T. (1991). Qualitative and quantitative analysis of phenolic constituents from Orthsoiphon arLstatus. Planta Medlca, 57: 176-180. Takeda, R., Hasegawa, J. and Sinozaki, M. (1990). The first isolation of lignans, megacerotonic and anthocerotonic acid, from non-vascular plants, Anthocerotae (hornworts). Tetrahedron Letters, 31: 4159-4162. Williamson, G., Day, A.J., Plumb, G.W. and Couteau, D. (2000). Human metabolic pathways of dietary flavonoids and cinnamates. Biochemlcal Society Transaction, 28: 16-22.
"This page is Intentionally Left Blank"
•
22 Green Tea: Molecular Targets in Glucose Uptake, Inflammation, and Insulin Signaling Pathways HEPING CAOl'*,.ANNE M. ROUSSEL 2,3 AND RICHARD A. ANDERSON l
ABSTRACT Obesity and related disorders including diabetes and cardiovascular diseases have been studied extensively, but the prevention and treatment of these health conditions remains inadequate. Green tea has anti-inflammatory, anti-diabetic, and anti-obesity activities, but the molecular mechanisms of these effects have not been fully elucidated. Quantitative real-time peR assays were used to investigate the effects of green tea extract on the expression of anti-inflammatory tristetraprolin family genes, pro-inflammatory genes, glucose transporter family genes, and insulin signaling pathway genes in liver and muscle of rats fed a high-fructose diet known to induce insulin resistance, oxidative stress, and inflammation. In this chapter, we review the experimental approaches used, the basal level expression of the various genes analyzed, and the transcriptional regulation of genes coding for the tristetraprolin family, glucose transporter family, pro-inflammatory and insulin signal transduction pathway components. The results show that antiinflammatory tristetraprolin mRNA levels are increased and proinflammatory tumor necrosis factor mRNA levels are reduced in the liver and skeletal muscle of rats fed a high-fructose diet given 1 g of green tea extract/kg of diet. Additional results suggest that green tea consumption 1. Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research
Center, Agricultural Research Service, US Department of Agriculture, Beltsville, MD, USA. 2. INSERM, U884, Grenoble, F-38000, France. 3. LBFA, Universite Joseph Fourier, Grenoble I, F-38041, France. Presently: South Regional Research Center, USDA-ARS, New Orleans, LA 70124, USA. * Corresponding author: E-mail: [email protected];[email protected]
430
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
regulates gene expression in glucose uptake and insulin signaling pathways in the same rats. These animal studies suggest that intake of adequate amounts ofgreen tea can effect mRNA message levels for genes that regulate inflammation and glucose metabolism in the liver and skeletal muscle. Key words: Gene expression, glucose transporter, green tea, high-fructose diet, inflammation, insulin signaling pathway, metabolic syndrome, rat, tristetraprolin, tumor necrosis factor
INTRODUCTION The extensive studies of obesity and related disorders including diabetes and cardiovascular diseases have not led to generally effective prevention and treatment of these health conditions (Dandona et al., 2004; Stumvoll et al., 2005; Eckel et al., 2005; Hotamisligil, 2006; Greenberg & Obin, 2006). Diet plays an important role in disease development, and the diets widely consumed in developed countries may increase the incidence of diabetes (Carter et al., 1996). The combination of higher content of refined sugars and fats and lower intake of traditional herbs, spices, and other plant products may contribute to the higher incidences of diabetes and obesity in the U.S. (Gross et al., 2004). Drug treatment for obesity and diabetes is not feasible for the majority of people and alternative and inexpensive therapies need to be evaluated. Medicinal plants have been used for the treatment of diabetes and related disorders for thousands of years (Gray & Flatt, 1997). Herbal medicines are playing a role in the prevention of obesity and control of type 2 DM for people with elevated levels of blood glucose and glucose intolerance who have a greater risk of developing diabetes. Plant seeds, fruits, leaves, and bark contain polyphenolic compounds. These compounds are the end products of the plant flavonoid biosynthetic pathway and are used by plants for protection against predators (Dixon et al., 2005). Plant polyphenols are also widely present in the diet (Prior & Gu, 2005) and are believed to be important for human health (Yang et al., 2001; McKay & Blumberg, 2002; Rodriguez et al., 2006). Tea (Camellia sinensis) is a popular beverage worldwide. Recent studies indicate that tea has a wide range of effects on animal and human health. A number of studies have indicated that green tea has antiinflammatory properties (Dona et al., 2003). Tea has been reported to have beneficial effects in conditions such as collagen-induced arthritis (Haqqi et al., 1999), inflammatory bowel disease (Varilek et al., 2001) and carrageenan-induced paw edema (Das et al., 2002). However, the molecular mechanisms oftea's anti-inflammatory properties have not been completely elucidated.
Green Tea in Glucose Uptake, Inflammation & Insulin
431
Other studies indicate that tea can be used to prevent diabetes and obesity for animal and human health (Wolfram et al., 2006; Kao et al., 2006). Several studies suggest that green tea extract (GTE) mimics insulin action. First, rat epididymal adipocyte assays indicate that GTE has insulinpotentiating activity on utilization of glucose (Broadhurst et al., 2000; Anderson & Polansky, 2002), and that the predominant active ingredient is epigallocatechin gallate (EGCG) (Broadhurst et al., 2000; Anderson & Polansky, 2002), the major polyphenol in green tea. Second, green tea powder and its polyphenols decrease fasting plasma levels of glucose, insulin, triglycerides, and free fatty acids, and increase the insulin-stimulated glucose uptake in rats fed green tea extract and polyphenols for 12 weeks (Wu et al., 2004a). Third, EGCG inhibits ~-2-aminobicycle (2.2.1)-heptane-2carboxylic acid-stimulated insulin secretion and glutamate dehydrogenase (Li et al., 2006a). Fourth, EGCG induces forkhead transcription factor family o phosphorylation by a similar but not identical mechanism to insulin and insulin growth factor I (Anton et al., 2007). A recent study suggests, however, that green tea consumption does not improve blood glucose, lipid profiles, insulin resistance or serum adiponectin levels in patients with type 2 diabetes (Ryu et al., 2006). Clearly, more studies are needed to demonstrate tea's insulin-like activity and the underlining molecular mechanisms. A high-fructose diet has been used as a model for the study of insulin resistance, oxidative stress (Faure et al., 1999), and inflammation (Kelley et al., 2004). Green tea was shown to decrease fasting plasma levels of glucose, insulin, triglycerides, and free fatty acids, and increase the insulin-stimulated glucose uptake and glucose transporter 4 (GLUT4) protein in rats fed a fructose-rich diet for 12 weeks (Wu et al., 2004b). However, that study did not address more potential targets in the GLUT family and the insulin signal transduction pathway (Cao et al., 2007c). Overproduction of free radicals under oxidative stress is associated with inflammation. Proinflammatory tumor necrosis factor alpha (TNF-a) levels are increased in rat muscle by high-fructose diet, which is one of the determinants of insulin resistance in skeletal muscle (Togashi et al., 2000; Yamaguchi et al., 2005). A high-fructose diet was also shown to activate inflammatory pathways in the liver (Kelley et al., 2004). Since liver and muscle are two of the most insulin-responsive organs and anti-inflammatory tristetraprolin/zinc finger protein 36 (TTP/ZFP36) gene expression is induced by insulin (Lai et al., 1990), we hypothesized that TTP and/or its homologues might be involved in the inflammatory response induced by a high-fructose diet in rats, and that green tea might possess beneficial effects in response to inflammation. We used quantitative real-time PCR to investigate the effects of GTE on the expression of numerous genes (Table 1) including the antiinflammatory TTP family mRNAs and some of the pro-inflammatory mRNAs
432
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
known to be regulated by TTP family proteins, and the GLUT family (Shepherd & Kahn, 1999) and insulin signal transduction pathway family genes (Taha & Klip, 1999; Cao et ai., 2007c). In this chapter, we review the experimental approaches used, the basal level expression of the various genes analyzed, and the transcriptional regulation of genes in the TTP family, GLUT family, pro-inflammatory and insulin signaling pathway components. Our results show that TTP mRNA levels are increased and TNF mRNA levels are reduced in the liver and skeletal muscle of rats fed a high-fructose diet given 1 g of GTE/kg of diet (Cao et ai., 2007b). Results also suggest that green tea consumption regulates the expression of genes coding for the glucose uptake and insulin signaling pathway proteins in the same rats (Cao et ai., 2007a). These studies suggest that intake of adequate amounts of green tea can effect mRNA message levels for genes that regulate inflammation and glucose metabolism in the liver and skeletal muscle that could lead to palliative effects on inflammation induced by high fructose in the diet. Table 1. Categories of the molecular targets analyzed by PCR assays Category
mRNA targets
Anti-inflammation
TIP/ZFP36, ZFP36L1, ZFP36L2, ZFP36L3
Pro-inflammation Glucose transport
TNF, GM-CSF, COX2, HUR, VEGF GLUT1, GLUT2, GLUT3, GLUT4 INS1, INS2, INSR, IRS1, IRS2, AKT1, GRB2, IGF1, IGF2, IGFlR, IGF2R, GSK3B, GYS1, PIK3CB, PIK3R1, SHC1, SOSl
Insulin signaling
EXPERIMENTAL APPROACHES Animals, Fructose-rich Diet, and Green Tea Extract Male Wistar rats (6 weeks old, - 150 g) were housed individually in thermoformed polystyrene cages. The rats were kept under the conditions with a 12 h light: 12 h dark schedule, at 21 ± 1°C, and a relative humidity of 55%. All procedures were in accord with guidelines of the U.S. National Institutes of Health. The composition of the fructose-rich diet (60% fructose, w/w) (SAFE, 89290, Augis, France) is given in Table 2. Green tea extract (GTE) (Unilever France) was prepared by boiling leaves in hot water. The dried water-soluble extract was added to the diet at 1 or 2 g/kg of diet. GTE contained 12.75% (w/w) epigallocatechin-3-gallate (EGCG), 9.21% epigallocatechin (EGC), 3.73% epicatechin gallate (ECG), 2.4% epicatechin (EC), 5.94% caffeine, and 0.195% L-theanine (Table 2).
Green Tea in Glucose Uptake, Inflammation & Insulin
433
Table 2. Composition ofthe high-fructose diet and green tea extract used in the study High-fructose diet (SAFE, France) 65% (w/w) fructose 20% casein 5% corn oil 5% alphacel 3.5% mineral mix 1% vitamin mix 0.3% DL-methionine 0.2% choline bitartrate
Green tea extract (Unilever France) 12.75% (w/w) EGCG (epigallocatechin-3-gallatel 9.21% EGC (epigallocatechin) 3.73% ECG (epicatechin gallate) 2.4% EC (epicatechin) 5.94% caffeine 0.195% L-theanine
Experimental Design Rats were fed a standard Purina chow for one week before being randomly divided into three groups (10 rats/group). The first group was given the high-fructose diet that has been shown to induce insulin resistance, oxidative stress, and inflammation (diet control). The second group was given the high-fructose diet plus 1 g of GTE/kg diet (1 g tea) and the third group was given the high-fructose diet plus 2 g of GTE/kg diet (2 g tea). Rats were sacrificed after 6 weeks on the diet. Food intake and the body weight for rats fed the diet control, 1 g tea, and 2 g tea for 6 weeks were not significantly different. These data are in agreement with a previous report in which the same diet was used and a similar evolution in body weight was reported CBusserolles et al., 2003). The liver and skeletal muscle were removed from the rats, frozen in liquid nitrogen, and stored at -80°C.
Gene Expression Analysis Total RNA was isolated from rat liver and muscle using TRI zOL reagent according to the manufacturer's instructions (Invitrogen, Carlsbad, CA, USA). RNA integrity and concentrations were determined using RNA 6000 Nano Assay Kit and the Bioanalyzer 2100 according to the manufacturer's instructions (Agilent Technologies, Palo Alto, CA, USA). Total cDNA synthesis was performed using the ImProm-II Reverse Transcription System (Promega, Madison, WI). The primers and probes were designed using Primer Express software (Applied Biosystems, Foster City, CA, USA). The mRNA names, GenBank accession numbers, and amplicon sizes are described (Table 3). The sequences (5' to 3') ofthe forward primers, TaqMan probes (TET - BHQl), and reverse primers have been described (Cao et al., 2007 a; Cao et al., 2007b). Quantitative real-time PCR assays were performed using an ABI Prism 7700 real-time PCR instrument (Applied Biosystems) (Dawson et al., 2005). The double CT method of relative quantification was used to determine the fold changes in expression (Livak & Schmittgen, 2001). This was done by first normalizing the resulting threshold cycle (CT)
434
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
values ofthe target mRNAs to the C T values ofthe internal control RPL32 in the same samples. It was further normalized with the diet control (samples with only the high-fructose diet but without tea supplement). The fold change in expression was then obtained. Data were analyzed by SigmaS tat 3.1 software (Systat Software, Inc., Point Richmond, CA) using One Way Analysis of Variance (ANOVA). Multiple comparisons were performed with Duncan's Multiple Range Test. Values with different lower case and upper case letters displayed above the columns of the figures are significantly different at p
Accession No
Amplicon size
mRNA
Accession Amplicon size No
NM_011756 NM_OO7564 NM_OOlOO1806 NM_OO1009549 NM_013693 NM_011198 NM_OO9969 NM_OlO485 NM_OO1025250 NM_011697 NM_172086 M13979 NM_0l2879 NM_0l7102 NM_012751
70bp 60bp 77bp 70bp 66bp 106bp 71bp 69bp 68bp 83bp 66bp 123bp 80bp 112bp 87bp
AKT1 GRB2 GSK3B GYS1 INS 1 INS2 INSR IGF1 IGFlR IGF2 IGF2R IRSI IRS2 PIK3CB PIK3R1 SHC1 SOSl
NM_033230 NM_030846 NM_032080 XM_229128 NM_OO8386 NM_OO8387 NM_0l7071 NM_184052 NM_OlO513 NM_OlO514 NM_OlO515 NM_0l2969 AF050159 NM_053481 NM_0l3005 XM_216176 D83014
90bp 119bp 106bp 119bp 89bp lOObp 137bp 78bp 62bp 78bp 91bp 68bp 69bp 134bp 118bp 85 bp 104bp
BASAL GENE EXPRESSION LEVELS TTP Family mRNA Levels in Rat Liver and Muscle Four forms of the anti-inflammatory TTP family proteins exist in rats and mice (TTP/ZFP36, ZFP36L1, ZFP36L2, and ZFP36L3) (Blackshear, 2002; Blackshear et al., 2005). The relative expression levels of these genes are important for the evaluation of GTE effects in the tissues. The relative ratios of TTP family mRNAs in the liver and muscle are shown in Table 4. TTP and ZFP36L1 mRNAs are the major TTP family members in liver and muscle (Cao et al., 2007b). The relative levels of TTP, ZFP36L1, and ZFP36L2 mRNAs in liver are 100%, 148%, and 6%, and those in muscle
Green Tea in Glucose Uptake, Inflammation & Insulin
435
are 100%, 70%, and 15%, respectively (Table 4) (Cao et at., 2007b). TTP, ZFP36L1, and ZFP36L2 mRNA levels are more abundant in the liver than those in the muscle (Table 4) (Cao et at., 2007b). ZFP36L3 mRNA is too low to be detected by 50 cycles ofPCR in the liver or muscle (Cao et at., 2007b). As a comparison, ZFP36L3 mRNA can be readily detected in cultured mouse-derived 3T3-L1 adipocytes and RAW 264.7 macrophages (Cao et at., 2008a; Cao et at., 2008b). Table 4. Relative mRNA levels ofTTP and GLUT family genes in rat liver and muscle [modified from (Cao et al., 2007a; Cao et al., 2007b)] Expression ratio (relative to TTP or GLUTl) (Fold)
Expression ratio (relative to liver) (Fold)
TTPtrIS11JZFP36 ZFP36LltrISllB ZFP36L2trISllD ZFP36L3 TTPtrIS11JZFP36
1.00 1.48 0.06 0.00 1.00
1.00 1.00 1.00
ZFP36LltrISllB ZFP36L2trISllD ZFP36L3
0.70 0.15
Tissue
mRNA
Liver
Muscle
Liver
Muscle
0.21 0.07
0.50
GLUTllSLC2A1 GLUT2ISLC2A2 GLUT3/SLC2A3
0.00 1.0 32.0 0.02
1.0 1.0 1.0
GLUT4ISLC2A4 GLUTllSLC2A1 GLUT2ISLC2A2
0.01 1.0 0.003
1.0 0.1 0.00001
GLUT3/SLC2A3
1.6
9
GLUT4ISLC2A4
294
4705
TTP Family-related mRNA Levels in Rat Liver and Muscle TNF and cyclooxgenase-2 (COX2) mRNA levels are very low in both liver and muscle (Cao et at., 2007b), and TNF mRNA levels in the liver are about 2-fold those in the muscle (Cao et at., 2007b). COX2 mRNA levels in the liver are about 40% of those in the muscle (Cao et at., 2007b). The expression levels ofHu antigen R (HuR), vascular endothelial growth factor a (VEGFA) and VEGFB in the liver are about 2.5-fold, 2-fold, and 25% of those in the muscle, respectively (Cao et at., 2007b). Granulocytemacrophage colony-stimulating factor (GM-CSF) mRNA is undetectable by 50 cycles ofPCR in the liver or muscle (Cao et at., 2007b). As a positive control for the assay, GM-CSF mRNA is detected in cultured RAW264.7 cells (Cao et at., 2008a).
436
Compo Bio. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
Glucose Transporter Family mRNA Levels in Rat Liver and Muscle The predominant glucose transporters in mammals are GLUTl, GLUT2, GLUT3, GLUT4, and GLUT5 (Shepherd & Kahn, 1999). The relative ratio of GLUT family mRNAs in the liver and muscle are shown in Table 4. GLUT2 is the major GLUT mRNA in the liver and GLUT4 is the major one in the muscle (Cao et al., 2007a). The relative mRNA levels of GLUTl, GLUT2, GLUT3, and GLUT4 in liver are 1-, 32-, 0.02-, and O.OI-fold, and those in muscle are 1-, 0.003-, 1.6-, and 294-fold, respectively (Table 4) (Cao et al., 2007a). GLUTl, GLUT2, GLUT3, and GLUT4 mRNAs in the muscle are 0.1-, 0.00001-,9-, and 4705-fold of those in the liver, respectively (Table 4) (Cao et al., 2007 a).
Insulin Signaling Pathway mRNA Levels in Rat Liver and Muscle Numerous components exist in the insulin signal transduction pathway (Taha & Klip, 1999). We have analyzed the mRNA levels of the following genes (refer to abbreviation list for gene/mRNA abbreviations) coding for insulin signaling pathway components (INSl, INS2, INSR, IRSl, IRS2, AKTl, GRB2, IGFl, IGF2, IGFIR, IGF2R, GSK3B, GYSl, PIK3CB, PIK3Rl, SHCl, SOSI) in liver and muscle of rats fed a high-fructose diet (Cao et a!., 2007a). AKTl, GRB2, GSK3B, INSR, IGF2R, IRSl, IRS2, PIK3CB, PIK3Rl, and SHCl mRNAs are more abundant in the liver than those in the muscle, whereas GYSl, INS2, IGF2, and IGFIR mRNAs are more abundant in the muscle than those in the liver (Cao et al., 2007a). INSl, INS2, IGFl, or IGF2 mRNAs is either undetectable or expressed at very low levels (Cao et al.,2007a).
REGULATION OF INFLAMMATION
GENE
EXPRESSION
INVOLVED
IN
Green Tea Increases Anti-inflammatory TTP mRNA Levels in Rat Liver and Muscle Green tea extract (1 g solid/kg diet) increases TTP mRNA levels by 50% and 140% in the liver and skeletal muscle, respectively, but does not have significant effects on TTP homologues ZFP36Ll or ZFP36L2 mRNA levels in the liver (Fig lA) (Cao et al., 2007b). However, GTE (2 g solid/kg diet) does not have significant effects on TTP, ZFP36Ll or ZFP36L2 mRNA levels in either tissue (Cao et al., 2007b). The percentage increases ofTTP mRNA in the muscle (Fig IE) are greater than those in the liver (Fig lA). Because TTP mRNA levels in the liver are about 5-fold higher than those in the muscle (Table 4), the net increases of TTP mRNA levels in the liver are more than those in the muscle (Cao et al., 2007b).
Green Tea in Glucose Uptake, Inflammation & Insulin
a a a
A B A
200 ,-- - -
437
a a a
180 ~ 180
~ 14O Gi 120 1; 100 c( 80 ~ 60 E 40 20
o
TTP (ZFP36)
ZFP36L1 ZFP36L2 TTP family mRNAs In rat liver
A B A
a a a
ZFP36L3
a a a
200 ~---------------------------------------
.-180 !160 1! 140 .i 120 ~ 100 a:: 80 E 60 40 20
o
TTP (ZFP36)
ZFP36L1
ZFP36L2
ZFP36L3
TTP family mRNA. In rat liver
Fig 1. Green tea extract effects on TIP family mRNA levels in rat liver and muscle. RNAs were isolated from livers and muscles of rats with metabolic syndrome induced by a high-fructose diet and transcribed into cDNAs. RNA-derived cDNAs (25 ng) were used for quantitative RT-PCR assays. The results represent the percentage means and the standard deviations from 5-8 samples. Values with different upper case and lower case letters displayed above the columns of the figure are significantly different at p
Green T e a Decrease s Pro-inflammatory TNF mRNA Le vels in Rat Liver and Decreases TNF and COX2 mRNA Levels in Rat Muscle The increases ofthe anti-inflammatory TTP mRNA levels with GTE suggest that green tea may have destabilizing effects on pro-inflammatory AUrich (ARE)-containing mRNAs such as TNF, GM-CSF, and COX2 mRNAs, whose stability are known to be destabilized by TTP (Carballo et al., 1998; Carballo et al., 2000; Sawaoka et al., 2003). PCR assays demonstrate that GTE at 1 g decreases TNF mRNA levels by 30% but does not have significant effects on COX2 mRNA levels in the liver (Fig 2A) (Cao et al., 2007b). PCR analyses also show that GTE at 1 g decreases both TNF and COX2 mRNA levels in the skeletal muscle by 30% and 40%, respectively (Fig 2B) (Cao et al., 2007b). However, GTE at 2 g does not have significant effects on TNF
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
438
or COX2 mRNA levels in the liver or muscle (Cao et al., 2007b). GM-CSF mRNA was undetectable by the PCR assays in the liver or muscle of rats treated with or without the tea supplement. 200 180 ~ 160 ~ 140 Qj > 120 .!! 100 « z 80 a: 60 E 40 20 0
a
a
b
a
a
a
• Diet control • 1 9 GTE/kg diet 0 2 9 GTElkg diet
~
TNF
GM-CSF
COX2
Pro-inflammatory mRNAs in rat liver
~
~
~
Qj
>
.!!
« z
a:
E
200 180 160 140 120 100 80 60 40 20 0
A
a
A
B
b
a
• Diet control • 1 9 GTE/kg diet 0 2 9 GTElkg diet
TNF
GM-CSF
COX2
Pro-inflammatory mRNAs in rat muscle
Fig 2. Green tea extract effects on TNF, GM-CSF, and COX2 mRNA levels in rat liver and muscle. RNA isola tion, cDNA synthesis , and real-time PCR assays are described in Fig 1 legend [modified from (Cao et al., 2007b))
Green Tea Effects on HuR and VEGF mRNA Levels in Rat Liver and Muscle
In contrast to TIP, HuR is known to be a stabilizing protein for some AREcontaining mRNAs (Atasoy et al., 1998; Raghavan et al., 2001; Katsanou et al., 2005). GTE at 1 g does not significantly affect HuR mRNA levels in the liver or muscle (Cao et al. , 2007b). However, GTE at 2 g increases the mRNA levels by 40% in the liver but not in the muscle (Cao et al. , 2007b). Similar to TNF and COX2 mRNAs, VEGF mRNA also contains ARE in the mRNA, which are destabilized by TIP family proteins (Ciais et al., 2004; Essafi-Benkhadir et al., 2007; Suswam et al., 2008). PCR assays demonstrate that GTE at 1 or 2 g does not have significant effects on VEGFA or VEGFB mRNA levels in the liver or muscle of rats fed the high-fructose diet (Cao et al. , 2007b).
Green Tea in Glucose Uptake, Inflammation & Insulin
439
REGULATION OF GENE EXPRESSION INVOLVED IN GLUCOSE UPTAKE AND INSULIN SIGNALING PATHWAY Green Tea Regulates the Expression of Genes Involved in Glucose Uptake Green tea extract (1 g GTE/kg diet) increases GLUTI and GLUT4 mRNA levels by 111% and 165%, respectively, but does not have significant effects on GLUT2 or GLUT3 mRNA levels in the liver (Fig 3A) (Cao et al., 2007a). GTE (2 g GTE/kg diet) increases GLUT4 mRNA levels by 92%, but does not have significant effects on GLUTl, GLUT2, or GLUT3 mRNA levels in the liver (Fig 3A) (Cao et al., 2007a). However, GTE (1 g GTE/kg diet) does not alter the mRNA levels of GLUT 1, GLUT2, GLUT3, or GLUT4 in the muscle (Fig 3B) (Cao et al., 2007a). GTE (2 g GTE/kg diet) increases GLUT2 and GLUT4 mRNA levels by 81% and 38% over the diet control, respectively, but does not have significant effects on GLUTI or GLUT3 mRNA levels in the muscle (Fig 3B) (Cao et al., 2007a). 350
....... 300 :.? e.....
A B A
a a
a
a a a
ABC
• Control diet • 1 9 GTE/kg diet 02 9 GTE/kg diet
4i 250 > .!! 200 c:(
z
150
a: E 100 50 0 GLUT1
GLUT2
GLUT3
GLUT4
GLUT family mRNAs In rat liver 300 250
a
a a
a a b
a a a
a ab a
. Control diet . 1 9 GTElkg diet 02 9 GTElkg diet
~ e..... 200
4i > .!!
150
c:(
z
a:
E
100 50 0 GLUT1
GLUT2
GLUT3
GLUT4
GLUT family mRNAs in rat muscle
Fig 3. Green tea extract effects on GLUT family mRNA levels in rat liver and muscle. RNA isolation, cDNA synthesis, and real-time PCR assays are described in Fig 1 legend [modified from (Cao et al., 2007a)]
Compo B io. Nat. Prod., Vol. 2 -Efficacy, Safety & Clinical Evaluation I
440
Green Tea R egulates the Expression of Genes Involved in Insulin Signaling Green tea extract (1 g GTE/kg diet) increases glycogen synthase kinase 3 beta (GSK3B) and insulin receptor substrate 2 (IRS2) mRNA levels by 34% and 55%, respectively (Fig 4A) (Cao et ai., 2007a), but does not have significant effects on AKT1, GRB2, GYS1, INS1, INS2, INSR, IGF1R, IGF2R, IRS1, PIK3R1, SHC1, or SOSl mRNA levels in the liver (Cao et ai., 2007 a). GTE (2 g GTE/kg diet) increases GSK3B and phosphatidylinositol 3-kinase, catalytic, beta (PIK3CB) mRNA levels by 30% and decreases Src homology 2 domain-containing transforming protein 1 (SHC1) mRNA levels by 63% (Fig 4A) (Cao et ai., 2007a), but does not have significant effects on AKT1, GRB2, GYS1, INS1, INS2, INSR, IGF1R, IGF2R, IRS1, IRS2, PIK3R1, or SOSl mRNA levels in the liver (Cao et ai., 2007a). In the muscle, GTE (1 a a b a b a a ab a a a b 250 ,-------------------------------------, . Control diet .1 9 GTElkg diet 02 9 GTElkg diet
~
150
~
< z 100
IX:
E
50
o GSK3B IRS2 PIK3CB SHC1 Insulin signaling pathway mRNAs in rat liver
250
a
b
ab
a
ab
b
a
ab
b
• Control diet . 1 9 GTElkg diet 0 2 9 GTElkg diet
200
e
..• >
< z
150 100
IX:
E
50 0
IRS1
SHC1
SOS1
Insulin signaling pathway mRNAs in rat muscle
F ig 4. Green tea extract effects on insulin signaling pathway mRNA levels in rat liver and muscle. RNA isolation, cDNA synthesis, and real-time PCR assays are described in Fig 1 legend [modified from (Cao et al., 2007a))
Green Tea in Glucose Uptake, Inflammation & Insulin
441
g GTE/kg diet) increases IRS1 mRNA levels by 84% (Fig 4B) (Cao et al., 2007a), but does not have significant effects on AKT1, GRB2, GSK3B, GYS1, INS1, INS2, INSR, IGF1R, IGF2R, IRS2, PIK3CB, PIK3R1, SHC1 or SOSl mRNA levels (Cao et al., 2007a). GTE (2 g GTE/kg diet) increases SHC1 and Son of sevenless 1 (SOSl) mRNA levels by 60% and 49%, respectively (Fig 4B) (Cao et al., 2007a), but does not have significant effects on AKT1, GRB2, GSK3B, GYS1, INS1, INS2, INSR, IGF1R, IGF2R, IRS1, IRS2, PIK3CB, or PIK3R1 mRNA levels in the muscle (Cao et al., 2007a).
DISCUSSION Plants have been used for the treatment of diabetes for thousands of years (Gray & Flatt, 1997), however, no investigation at the molecular level was substantially performed to support this practice. Several studies indicate that tea extract and its major polyphenol, EGCG, has insulin-potentiating activity in in vitro and in animal models (Broadhurst et al., 2000; Anderson & Polansky, 2002; Wu et al., 2004b; Anderson et al., 2005; Li et al., 2006b; Anton et al., 2007) and that green tea has anti-inflammatory properties (Dona et al., 2003). We recently analyzed the effects of a green tea polyphenolic extract on the mRNA levels of glucose transporter family, insulin signal transduction pathway family, anti- and pro-inflammatory family genes in liver and muscle of rats fed a high-fructose diet known to induce insulin resistance, oxidative stress (Faure et al., 1999), and inflammation in rats (Kelley et al., 2004). One of the major findings is that GTE increases anti-inflammatory TTP and decreases pro-inflammatory TNF gene expression in liver and muscle of rats fed a high-fructose diet (Cao et al., 2007b). Recent investigations have established a mechanism for the regulation of inflammatory responses at the post-transcriptional level by TTP family proteins (Blackshear, 2002; Anderson et al., 2004). TIP binds ARE in some mRNAs and destabilizes those transcripts encoding proteins such as TNFa (Carballo et al., 1998; Lai et al., 1999; Cao et al., 2003; Cao, 2004), GMCSF (Carballo et al., 2000; Carballo et al., 2001), and COX2 (Sawaoka et al., 2003). The mRNAs encoding TNF-a and GM-CSF are stabilized in TIP knockout mice and in cells derived from them (Carballo et al., 1998; Carballo et al., 2000). Excessive secretion of these cytokines in TTP knockout mice results in a severe systemic inflammatory response including arthritis, autoimmunity, and myeloid hyperplasia (Taylor et al., 1996; Phillips et al., 2004). Up-regulation of TTP reduces inflammatory responses in macrophages (Sauer et al., 2006). These lines of evidence support the conclusion that TTP is an anti-inflammatory protein. Therefore, agents that induce TIP gene expression may have potential therapeutic value for the prevention and/or treatment of inflammation-related diseases. Our results show that GTE increases TIP and decreases TNF gene expression in rats fed a high-fructose diet known to induce inflammation, suggesting
442
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
that part of the mechanism of tea's anti-inflammatory effects may involve TTP at the post-transcriptional level. The other major finding is that GTE increases GLUT4 mRNA levels in rat liver and muscle of rats fed a high-fructose diet (Cao et al., 2007a). The increased GLUT4 mRNA level in the muscle may be physiologically significant for the following reasons: 1) GLUT4 is the major GLUT family mRNAs in the muscle (Table 4), 2) GLUT4 protein is the insulinresponsive glucose transporter in the muscle and adipose tissue (Shepherd & Kahn, 1999), 3) the amount of GLUT4 protein is decreased in obese subjects (Garvey et al., 1991), 4) insulin increases GLUT4 mRNA and protein levels in fetal rat brown adipocytes (Valverde et al., 1999; Hernandez et al., 2003) and increases GLUT4 protein levels in 3T3-F442A adipocytes (Yu et al., 2001), and 5) the amount of GLUT4 protein is increased in fructose-fed rats with green tea powder supplementation resulting in amelioration of fructose-induced insulin resistance (Wu et al., 2004b). The increased GLUT4 mRNA levels with GTE suggest a positive effect of these polyphenolic compounds on the long-term regulation of glucose transport. The significance of the increased levels of GLUTI and GLUT4 mRNAs in the liver and GLUT2 mRNA in the muscle is not clear because these GLUT mRNAs are the minor forms in their respective rat tissues (Table 4). Our results also show that GTE consumption affects the expression of other genes coding for key proteins in the insulin-signaling pathway. GTE increases GSK3B, IRS2 and PIK3CB, but decreases SHCl mRNA levels in the liver, and GTE increases IRSl, SHCl, and SOSI mRNA levels in the muscle (Cao et al., 2007a). However, GTE consumption exhibits none or minimal effects on the other mRNA levels in the liver or muscle, including INSl, INS2, INSR, AKTl, GRB2, IGFl, IGF2, IGFIR, IGF2R, GYSl, and PIK3Rl mRNAs (Cao et al., 2007a). The small effects of GTE consumption on mRNA levels in the insulin signaling pathway in our studies are in agreement with previous observations from other studies showing that the effects of insulin on the expression of a number of genes in the insulin signaling pathway are minimal in mouse 3T3-Ll adipocytes (Wang et al., 2006; Cao et al., 2008b). The plausible reason for the small effects of GTE and insulin on mRNA levels in the insulin signal transduction pathway in rat liver and muscle, and cultured mouse-derived 3T3-Ll adipocytes, is probably due to the functional effects of insulin signal transduction mainly through protein phosphorylation. It is also possible that the effectiveness of GTE is dependent upon factors such as the timing and dosages, and the physiological status of the target cells. Additional studies are needed to determine a useful role for GTE and its components in improving the negative health effects of glucose intolerance, insulin resistance, diabetes, and obesity.
Green Tea in Glucose Uptake, Inflammation & Insulin
443
Food intake for rats fed 1 g GTElkg diet is 20.5 g/d. On a per kg basis, the rats consuming 1 g of GTE per day consume approximately 60 mglkg body weight per day. Humans consuming 1 cup of tea made from a tea bag containing 2 g of solids consume roughly 900 mg of tea solids or approximately 12.8 mglkg. On a per kilogram basis, 1 g GTElkg diet used in our study is equivalent to humans drinking approximately 5 cups of tea per day. The reason(s) why GTE at 2 glkg of diet does not produce more TIP mRNA levels than 1 g of GTElkg diet in the liver is not clear. The effects of GTE on insulin levels in the plasma showed a similar pattern (Anderson et al., 2005). Additional dose response studies of GTE and its components are needed.
CONCLUSIONS We have investigated the effects of GTE on the expression of multiple genes coding for inflammatory factors, GLUT family and components in the insulin signal transduction pathway (Fig 5). Quantitative real-time peR demonstrate that anti-inflammatory TIP mRNA levels are increased significantly and pro-inflammatory TNF mRNA levels are reduced in the liver and skeletal muscle of rats fed a high-fructose diet and given 1 g GTElkg in the diet.
Fig 5. A working model for green tea extract in inflammation, glucose uptake, and insulin signaling pathway leading to potential beneficial effects. This model is modified from a model for cinnamon polyphenol effects (Cao et al., 2007c). The effects of green tea polyphenol extract on TTP and TNF gene expression are described in Figs 1,2 (Cao et al., 2007b), and those on GLUT4 and GSK3B gene expression are described in Figs 3, 4 (Cao et al., 2007a) ("+" and "-"represent positive and negative effects, respectively) [from (Cao et al., 2007a))
444
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Additional results suggest that GTE consumption regulates gene expression in glucose uptake and insulin signaling pathways in the same rats. These findings support plausible reasons for drinking adequate amounts of green tea for improved health consequences.
ABBREVIATIONS ANOVA: one way analysis of variance; ARE: AU-rich element; AKT1: thymoma viral proto-oncogene 1; COX-2/PTGS2: cyclooxgenase-21 prostaglandin-endoperoxide synthase 2; EGCG: Epigallocatechin-3-gallate; GLUT: glucose transporter; GM-CSF/CSF2: granulocyte-macrophage colonystimulating factor; GRB2: growth factor receptor bound protein 2; GSK3B: glycogen synthase kinase 3 beta; GTE: green tea extract; GYS1: glycogen synthase 1; HuRlELAVL1: Hu antigen Rlembryonic lethal, abnormal visionlike 1; IGF1: insulin-like growth factor 1; IGF1R: insulin-like growth factor I receptor; IGF2: insulin-like growth factor 2; IGF2R: insulin-like growth factor 2 receptor; INS1: insulin I; INS2: insulin II; INSR: insulin receptor; IRS1: insulin receptor substrate 1; IRS2: insulin receptor substrate 2; PIK3CB: phosphatidylinositol 3-kinase, catalytic, beta; PIK3R1: phosphatidylinositol 3-kinase, regulatory subunit 1; SHC1: Src homology 2 domain-containing transforming protein 1; SOSl: Son of sevenless 1; RPL32: ribosomal protein L32; TNF: tumor necrosis factor; TTP: tristetraprolin; VEGF: vascular endothelial growth factor; ZFP36: zinc finger protein 36; ZFP36L: ZFP36-like. ACKNO~DGEMENTS
This work was presented at the Fourth International Scientific Symposium on Tea & Human Health, U.S. Department of Agriculture Jefferson Auditorium, Washington, DC, September 18, 2007. The original work was supported in part by USDA-ARS Human Nutrition Research Program. This work is part of the Scientific Cooperative Convention Agreement (# 581235-4N-F033) between the Human Nutrition Research Center (Beltsville, MD, USA) and the Joseph Fourier University (Grenoble, France). We thank Harry D. Dawson and Meghan A. Kelly for technical assistance and Joseph F. Urban Jr. for helpful comments on the manuscript.
REFERENCES Anderson, P ., Phillips, K., Stoecklin, G. and Kedersha, N . (2004). Post-transcriptional regulation of proinflammatory proteins. Journal of Leukocyte Biology, 76: 42-47. Anderson, R.A. , Hininger, 1., Coves, S. and Roussel, A.M. (2005). Tea increases insulin sensitivity and decreases oxidative stress in rats with metabolic syndrome. Proceedings of the 18th International Congress of Nutrition , pp. 1.3.1. Anderson, R.A. and Polansky, M.M. (2002). Tea enhances insulin activity. Journal of Agricultural and Food Chemistry, 50: 7182-7186.
Green Tea in Glucose Uptake, Inflammation & Insulin
445
Anton, S., Melville, L. and Rena, G. (2007). Epigallocatechin gallate (EGCG) mimics insulin action on the transcription factor FOXOla and elicits cellular responses in the presence and absence of insulin. Cellular Signaling, 19: 378-383. Atasoy, U., Watson, J., Patel, D. and Keene, J. (1998). ELAV protein HuA (HuR) can redistribute between nucleus and cytoplasm and is upregulated during serum stimulation and T cell activation. Journal of Cell SCience, 111: 3145-3156. Blackshear, P.J. (2002). Tristetraprolin and other CCCH tandem zinc-finger proteins in the regulation of mRNA turnover. Biochemical Society Transactions, 30: 945-952. Blackshear, P.J., Phillips, RS., Ghosh, S., Ramos, S.B., Richfield, E.K. and Lai, W.S. (2005). Zfp3613, a rodent X chromosome gene encoding a placenta-specific member of the tristetraprolin family of CCCH tandem zinc finger proteins. Bwlogy of Reproduction, 73: 297-307. Broadhurst, C.L., Polansky, M.M. and Anderson, RA. (2000). Insulin-like biological activity of culinary and medicinal plant aqueous extracts in vitro. Journal of Agricultural and Food Chemistry, 48: 849-852. Busserolles, J., Gueux, E., Rock, E., Demigne, C., Mazur, A. and Rayssiguier, Y. (2003). Oligofructose protects against the hypertriglyceridemic and pro-oxidative effects of a high fructose diet in rats. Journal of Nutrition, 133: 1903-1908. Cao, H. (2004). Expression, purification, and biochemical characterization of the antiinflammatory tristetraprolin: a zinc-dependent mRNA binding protein affected by posttranslational modifications. Bwchemistry, 43: 13724-13738. Cao, H., Dzineku, F. and Blackshear, P.J. (2003). Expression and purification of recombinant tristetraprolin that can bind to tumor necrosis factor-a mRNA and serve as a substrate for mitogen-activated protein kinases. Archives of BiochemIstry and Biophysics, 412: 106-120. Cao, H., Hininger-Favier, 1., Kelly, M.A., Benaraba, R, Dawson, H.D., Coves, S., Roussel, A.M. and Anderson, RA. (2007a). Green tea polyphenol extract regulates the expression of genes involved in glucose uptake and insulin signaling in rats fed a high fructose diet. Journal of Agricultural and Food Chemistry, 55: 6372-6378. Cao, H., Kelly, M.A., Kari, F., Dawson, H.D., Urban, J.F. Jr. Coves, S., Roussel, A.M. and Anderson, RA. (2007b). Green tea increases anti-inflammatory tristetraprolin and decreases pro-inflammatory tumor necrosis factor mRNA levels in rats. Journal of Inflammatwn (London), 4: 1. Cao, H., Polansky, M.M. and Anderson, RA. (2007c). Cinnamon extract and polyphenols affect the expression of tristetraprolin, insulin receptor, and glucose transporter 4 in mouse 3T3-L1 adipocytes. Archives of Biochemistry and Biophysics, 459: 214-222. Cao, H., Urban, J.F., Jr. and Anderson, RA. (2008b). Insulin increases tristetraprolin and decreases VEGF gene expression in mouse 3T3-L1 adipocytes. Obesity (Silver. Sprmg), 16: 1208-1218. Cao, H., Urban, J.F. Jr. and Anderson, RA. (2008a). Cinnamon polyphenol extract affects immune responses by regulating anti- and proinflammatory and glucose transporter gene expression in mouse macrophages. Journal of Nutrition, 138: 833-840. Carballo, E., Cao, H., Lai, W.S., Kennington, E.A., Campbell, D. and Blackshear, P.J. (2001). Decreased sensitivity of tristetraprolin-deficient cells to p38 inhibitors suggests the involvement of tristetraprolin in the p. 38 signaling pathway. Journal of Bwlogical Chemistry, 276: 42580-42587. Carballo, E., Lai, W.S. and Blackshear, P.J. (2000). Evidence that tristetraprolin is a physiological regulator of granulocyte-macrophage colony-stimulating factor messenger RNA deadenylation and stability. Blood, 95: 1891-1899. Carballo, E., Lai, W.S. and Blackshear, P.J. (1998). Feedback inhibition of macrophage tumor necrosis factor-a production by tristetraprolin. Science, 281: 1001-1005. Carter, J.8., Pugh, J.A. and Monterrosa, A. (1996). Non-insulin-dependent diabetes mellitus in minorities in the United States. Annals of Internal Medicine, 125: 221-232. Ciais, D., Cherradi, N., Bailly, S., Grenier, E., Berra, E., Pouyssegur, J., LaMarre, J. and Feige, J.J. (2004). Destabilization of vascular endothelial growth factor mRNA by the zinc-finger protein TIS11b. Oncogene, 23: 8673-8680.
446
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Dandona, P., Aljada, A. and Bandyopadhyay, A. (2004). Inflammation: the link between insulin resistance, obesity and diabetes. Trends in Immunology, 25: 4-7. Das, M., Sur, P., Gomes, A., Vedasiromoni, J.R and Ganguly, D.K (2002). Inhibition of tumour growth and inflammation by consumption of tea. Phytotherapy Research, 16: S40-S44. Dawson, H.D., Beshah, E., Nishi, S., Solano-Aguilar, G., Morimoto, M., Zhao, A., Madden, KB., Ledbetter, T.K, Dubey, J.P., Shea-Donohue, T., Lunney, J.K and Urban, J.F. Jr. (2005). Localized multigene expression patterns support an evolving ThllTh2-like paradigm in response to infections with Toxoplasma gondii and Ascaris suum. Infection and Immunity, 73: 1116-1128. Dixon, RA., Xie, D.Y. and Sharma, S.B. (2005). Proanthocyanidins-a final frontier in flavonoid research? New Phytology, 165: 9-28. Dona, M., Dell'Aica, 1., Calabrese, F., Benelli, R, Morini, M., Albini, A. and Garbisa, S. (2003). Neutrophil restraint by green tea: inhibition of inflammation, associated angiogenesis, and pulmonary fibrosis. Journal of Immunology, 170: 4335-4341. Eckel, RH., Grundy, S.M. and Zimmet, P.Z. (2005). The metabolic syndrome. Lancet, 365: 1415-1428. Essafi-Benkhadir, K, Onesto, C., Stebe, E., Moroni, C. and Pages, G. (2007). Tristetraprolin inhibits ras-dependent tumor vascularization by inducing VEGF mRNA degradation. Molecular Biology of the Cell, 18: 4648-4658. Faure, P., Rossini, E., Wiernsperger, N., Richard, M.J., Favier, A. and Halimi, S. (1999). An insulin sensitizer improves the free radical defense system potential and insulin sensitivity in high fructose-fed rats. Diabetes, 48: 353-357. Garvey, W.T., Maianu, L., Huecksteadt, T.P., Birnbaum, M.J., Molina, J.M. and Ciaraldi, T.P. (1991). Pretranslational suppression of a glucose transporter protein causes insulin resistance in adipocytes from patients with non-insulin-dependent diabetes mellitus and obesity. Journal of Clinical Investigatwn, 87: 1072-1081. Gray, A.M. and Flatt, P.R (1997). Pancreatic and extra-pancreatic effects of the traditional anti-diabetic plant, Medicago satwa (lucerne). British Journal of Nutrition, 78: 325-334. Greenberg, A.S. and Obin, M.S. (2006). Obesity and the role of adipose tissue in inflammation and metabolism. American Journal of Clinical Nutrition, 83: 461S465S. Gross, L.S., Li, L., Ford, E.S. and Liu, S. (2004). Increased consumption of refined carbohydrates and the epidemic of type 2 diabetes in the United States: an ecologic assessment. American Journal of Clinical Nutrition, 79: 774-779. Haqqi, T.M., Anthony, D.D., Gupta, S., Ahmad, N., Lee, M.s., Kumar, G.K and Mukhtar, H. (1999). Prevention of collagen-induced arthritis in mice by a polyphenolic fraction from green tea. Proceedmgs of the National Academy of Sciences of USA, 96: 4524-4529. Hernandez, R, Teruel, T. and Lorenzo, M. (2003). Insulin and dexamethasone induce GLUT4 gene expression in foetal brown adipocytes: synergistic effect through CCAAT/enhancerbinding protein alpha. Biochemical Journal, 372: 617-624. Hotamisligil, G.S. (2006). Inflammation and metabolic disorders. Nature, 444: 860-867. Kao, Y.H., Chang, H.H., Lee, M.J. and Chen, C.L. (2006). Tea, obesity, and diabetes. Molecular Nutrition and Food Research, 50: 188-210. Katsanou, V., Papadaki, 0., Milatos, S., Blackshear, P.J., Anderson, P., Kollias, G. and Kontoyiannis, D.L. (2005). HuR as a negative posttranscriptional modulator in inflammation. Molecular Cell, 19: 777-789. Kelley, G.L., Allan, G. and Azhar, S. (2004). High dietary fructose induces a hepatic stress response resulting in cholesterol and lipid dysregulation. Endocrinology, 145: 548-555. Lai, W.S., Carballo, E., Strum, J.R, Kennington, E.A., Phillips, RS. and Blackshear, P.J. (1999). Evidence that tristetraprolin binds to AU-rich elements and promotes the deadenylation and destabilization of tumor necrosis factor alpha mRNA. Molecular and Cellular Biology, 19: 4311-4323. Lai, W.S., Stumpo, D.J. and Blackshear, P.J. (1990). Rapid insulin-stimulated accumulation of an mRNA encoding a proline-rich protein. Journal of Biological Chemistry, 265: 1655616563.
Green Tea in Glucose Uptake, Inflammation & Insulin
447
Li, C., Allen, A., Kwagh, J., Doliba, N.M., Qin, W., Najafi, H., Collins, H.W., Matschinsky, F.M., Stanley, C.A and Smith, T.J. (2006a). Green tea polyphenols modulate insulin secretion by inhibiting glutamate dehydrogenase. Journal of Biological Chemistry, 281: 10214-1022l. Li, R.W., Douglas, T.D., Maiyoh, G.K, Adeli, K and Theriault, AG. (2006b). Green tea leaf extract improves lipid and glucose homeostasis in a fructose-fed insulin-resistant hamster model. Journal of Ethnopharmacology, 104: 24-31. Livak, KJ. and Schmittgen, T.D. (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C (T)) Method. Methods, 25: 402-408. McKay, D.L. and Blumberg, J.B. (2002). The role of tea in human health: an update. Journal of American College of Nutrition, 21: 1-13. Phillips, K, Kedersha, N., Shen, L., Blackshear, P.J. and Anderson, P. (2004). Arthritis suppressor genes TIA-1 and TTP dampen the expression of tumor necrosis factor alpha, cydooxygenase 2, and inflammatory arthritis. Proceedings of the National Academy of Sciences of USA, 101: 2011-2016. Prior, RL. and Gu, L. (2005). Occurrence and biological significance of proanthocyanidins in the American diet. Phytochemistry, 66: 2264-2280. Raghavan, A, Robison, RL., McNabb, J., Miller, C.R, Williams, D.A. and Bohjanen, P.R (2001). HuA and tristetraprolin are induced following T cell activation and display distinct but overlapping RNA binding specificities. Journal of Biological Chemistry, 276: 4795847965. Rodriguez, S.K, Guo, W., Liu, L., Band, M.A, Paulson, E.K and Meydani, M. (2006). Green tea catechin, epigallocatechin-3-gallate, inhibits vascular endothelial growth factor angiogenic signaling by disrupting the formation of a receptor complex. International. Journal of Cancer, 118: 1635-1644. Ryu, O.H., Lee, J., Lee, KW., Kim, H.Y., Seo, J.A, Kim, S.G., Kim, N.H., Baik. S.H., Choi, D.S. and Choi, KM. (2006). Effects of green tea consumption on inflammation, insulin resistance and pulse wave velocity in type 2 diabetes patients. Diabetes Research and Clinical Pract~ce, 71: 356-358. Sauer, 1., Schaljo, B., Vogl, C., Gattermeier, 1., Kolbe, T., Muller, M., Blackshear, P.J. and Kovarik, P. (2006). Interferons limit inflammatory responses by induction oftristetraprolin. Blood, 107: 4790-4797. Sawaoka, H., Dixon, D.A., Oates, J.A. and Boutaud, O. (2003). Tristetraprolin binds to the 3'-untranslated region of cyclooxygenase-2 mRNA A polyadenylation variant in a cancer cell line lacks the binding site. Journal of Biological Chem~stry, 278: 13928-13935. Shepherd, P.R and Kahn, B.B. (1999). Glucose transporters and insulin action-implications for insulin resistance and diabetes mellitus. New England Journal of Med~cine, 341: 248257. Stumvoll, M., Goldstein, B.J. and van Haeften, T.W. (2005). Type 2 diabetes: principles of pathogenesis and therapy. Lancet, 365: 1333-1346. Suswam, E., Li, Y., Zhang, X., Gillespie, G.Y., Li, X., Shacka, J.J., Lu, L., Zheng, L. and King, P.H. (2008). Tristetraprolin down-regulates interleukin-8 and vascular endothelial growth factor in malignant glioma cells. Cancer Research, 68: 674-682. Taha, C. and Klip, A (1999). The insulin signaling pathway. Journal of Membrane Biology, 169: 1-12. Taylor, G.A., Carballo, E., Lee, D.M., Lai, W.S., Thompson, M.J., Patel, D.D., Schenkman, D.l., Gilkeson, G.S., Broxmeyer, H.E., Haynes, B.F. and Blackshear, P.J. (1996). A pathogenetic role for TNF -alpha in the syndrome of cachexia, arthritis, and autoimmunity resulting from tristetraprolin (TTP) deficiency. Immunity, 4: 445-454. Togashi, N., Ura, N., Higashiura, K, Murakami, H. and Shimamoto, K. (2000). The contribution of skeletal muscle tumor necrosis factor-alpha to insulin resistance and hypertension in fructose-fed rats. Journal of Hypertension, 18: 1605-1610. Valverde, AM., Navarro, P., Teruel, T., Conejo, R, Benito, M. and Lorenzo, M. (1999). Insulin and insulin-like growth factor I up-regulate GLUT4 gene expression in fetal brown adipocytes, in a phosphoinositide 3-kinase-dependent manner. Biochemical Journal, 337: 397-405.
448
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Varilek, G.W., Yang, F., Lee, E.Y., deVilliers, W.J., Zhong, J., Oz, H.S., Westberry, KF. and McClain, C.J. (2001). Green tea polyphenol extract attenuates inflammation in interleukin-2-deficient mice, a model of autoimmunity. Journal of Nutrition, 131: 2034-2039. Wang, P., Keijer, J., Bunschoten, A., Bouwman, F., Renes, J. and Mariman, E. (2006). Insulin modulates the secretion of proteins from mature 3T3-L1 adipocytes: a role for transcriptional regulation of processing. Diabetologia, 49: 2453-2462. Wolfram, S., Wang, Y. and Thielecke, F. (2006). Anti-obesity effects of green tea: from bedside to bench. Molecular Nutrition and Food Research, 50: 176-187. Wu, L.Y., Juan, C.C., Ho, L.T., Hsu, Y.P. and Hwang, L.S. (2004a). Effect of green tea supplementation on insulin sensitivity in Sprague-Dawley rats. Journal of Agriculture and Food Chemistry, 52: 643-648. Wu, L.Y., Juan, C.C., Hwang, L.S., Hsu, Y.P., Ho, P.H. and Ho, L.T. (2004b). Green tea supplementation ameliorates insulin resistance and increases glucose transporter IV content in a fructose-fed rat model. European Journal of Nutrition, 43: 116-124. Yamaguchi, K, Ura, N., Murakami, H., Togashi, N., Hyakukoku, M., Higashiura, K and Shimamoto, K (2005). Olmesartan ameliorates insulin sensitivity by modulating tumor necrosis factor-a and cyclic AMP in skeletal muscle. Hypertension Research, 28: 773-778. Yang, C.S., Landau, J.M., Huang, M.T. and Newmark, H.L. (2001). Inhibition of carcinogenesis by dietary polyphenolic compounds. Annual Review of Nutritwn, 21: 381-406. Yu, Z.W., Buren, J., Enerback, S., Nilsson, E., Samuelsson, L. and Eriksson, J.W. (2001). Insulin can enhance GLUT4 gene expression in 3T3-F442A cells and this effect is mimicked by vanadate but counteracted by cAMP and high glucose - potential implications for insulin resistance. Biochimica et Biophysica Acta, 1535: 174-185.
23 Euphorbious Plants as Molluscicides and Piscicides: A Review
ABSTRACT
Many aquatic snails act as vectors for the larvae of trematode and thereby cause a number of diseases. Two diseases carried by aquatic snails, fascioloiasis and schistosomiasis cause immense harm to man and his domestic animals. The WHO has tested thousands of synthetic compounds for the eradication of freshwater target snails. Though effective, these pesticides have so for not proved themselves to be entirely satisfactory. With a growing awareness of environmental pollution, which such compounds can cause, efforts are being made to find out molluscicidal and piscicidal products of plant origin. Presence of predatory fishes in fish culture pond is also a serious problem due to their growth and better utilization of cultured carp habitats and food. Due to their carnivorous nature they engulf the fingerlings ofcultured carps and adversely effect the aquaculture production. A number ofcompounds, such as saponins, tannins, miscellaneous alkaloids, alkenyl phenols, glycoalkaloids, flavonoids, sesquiterpenes lactones, terpenoids and phorbol esters present in various plants have been found to be toxic to target freshwater snails and predatory fishes. Key words : Euphorbiales, molluscicides, piscicides, snail
INTRODUCTION Although the exact number of existing molluscan species is still a matter of speculation, Abbott (1954) has estimated a total of about 1,10,000 living 1. Natural Products Laboratory, Department of Zoology, DDU, Gorakhpur University, Gorakhpur - 273 009 (U.P.) India. * Corresponding author: E-mail: [email protected]; singhajay_gkp@ rediffmail.com
450
Compo Bio. Nat. Prod., Vol. 2-Efficacy, Safety & Clinical Evaluation!
species, 80,000 amongst which are gastropods, 10,000 bivalves, and 5,000 belonging to the other three classes of mollusca. (Godan, 1983) on the other hand believe the number of living species is about 1,20,000. Terrestrial snails and slugs cause considerable damage to both cultivated and useful non-cultivated plants. The animals can make their appearance in any damp area, but damage can occur also during relatively dry weather. Along with slugs, terrestrial snails also cause considerable damage to vegetable gardens, agricultural crops and fruits orchards. Singh and Agarwal (1981), reported that Pila globosa an amphibious snails causes damage to paddy crops in northern part of India. In freshwater, the larvae of parasite trematodes also pass part of their life. Many aquatic snails act as vectors for the larvae of trematodes and there by, cause a number of diseases. Two diseases carried by aquatic snails, schistosomiasis and fascioliasis cause immense harm to man and his domestic animals (Bali et al., 1986; Yadav & Singh 2001, 2002, 2003; Yadav et al., 2004, 2005). Fasciola hepatica, the large liver fluke, common in sheep, cattle, goat and other herbivorous animals throughout the World. Froyed (1975), reported that about 21% cattle and 7% sheep were infected with liver fluke in Great Britain. In India, the freshwater snails Lymnaea acuminata and Indoplanorbis exustus are the intermediate host of Fasciola hepatica and Fasciola gigantica (Hyman, 1970; Singh & Agarwal, 1988a; Singh & Agarwal, 1992b; Yadav, 2000; Yadav & Singh, 2001, 2003), which causes immense harm to domestic animals of this country. Schistosomiasis is caused by Schistosoma, it is a devastating disease of mankind second only to malaria in its deleterious effect (Jobin, 1973). The control of harmful freshwater snails through synthetic pesticides has been review in detail by various workers (Ritchie, 1973; Godan, 1983; Agarwal & Singh, 1988; Singh & Agarwal, 1990; Singh et al., 1996).
SYNTHETIC MOLLUSCICIDES Metaldehyde The molluscicidal activity of metaldehyde is generally influenced by temperature and humidity. Its treatment affects the gastropods by two ways; first its irritant effect which causes gastropods to secretes large amount of mucus resulting in desiccation and secondary its neurotoxicity at high concentration (Stringer, 1946).
Niclosamide This is the standard molluscicides, active against all the stages of snails at 24h exposure up to 0.03 ppm. The niclosamide is not an irritant, as the snails treated with this compound, did not exhibit any symptoms like crawling, excessive production of mucus or quick contraction in to the shell (Khajuria & Bali, 1988).
Euphorbious Plants as Molluscicides and Piscicides
451
Carbamate and Organophosphate Compounds Carbamate and organophosphate (OP) compounds are esterase inhibiting neurotoxic ants , with acute cholinergic effects preceded by inhibition of acetylcholinesterase (AChE) (Matsumura, 1985).
Carbamate Pesticides such as carbaryl (Barry, 1969; Brar & Simwat, 1973; Singh & Agarwal, 1982), Aldicarb (Judge, 1969; Singh & Agarwal, 1981), Isolan (DaxI, 1971), Methiocarb (Rayner, 1975) and Mesurol (Smith & Boswell, 1970) have been used against various molluscan pests.
Organophosphate Pesticides which currently being used for the control of various species of gastropods pests are Zinophos (Barry, 1969), Phorate (Barry, 1969; Singh & Agarwal, 1983), Nuvan (Tripathi & Agarwal, 1998), Dimeton, Trichlorofon (Singh & Agarwal, 1981) and Karathane (Oteifa et al., 1975).
Synthetic Pyrethroids The pyrethroids act primarily on nerve membrane by changing it to Na+ and K+ permeability. This causes repetitive discharges of nerve at the synapse and neuromuscular junction (Narahashi, 1983). Adlung and Kauth (1956), however, found that a 5% oil emulsion of pyrethrum was quite effective against the freshwater snails, Radix auricularia, Lymnaea stagnalis and Physa fontinalis.
PLANT ORIGIN MOLLUSCICIDES With growing awareness of environmental pollution caused by synthetic molluscicides (Ritchie, 1973; Srivastava & Singh, 2001; Saravanan et al., 2003; Selvarani & Rajamanickam, 2003, Park et al., 2004), efforts are being made to find out molluscicides of plant origin. Being the product of biosynthesis, they are highly toxic and easily biodegradable in environment (Marston & Hostettman, 1987; Singh & Agarwal, 1992a; Singh et al., 1996). A continuous search for new classes ofmolluscicidal natural products is essential, so that problems of selectivity and low activity can be overcome. In addition, as wide a range as possible of structurally related compounds should be isolated (or synthesized) for structure-activity and mode of action studies. This paper review all the relevant information on Euphorbious plants, which is considered as one of the most promising known molluscicidal and piscicidal plants.
452
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Euphorbiales as Molluscicides (Table 1) Snails exposed to latex of Euphorbia royleana exhibited typical symptoms of nerve poisoning and death took place within 24 h. It was shown that the latex was an acetylcholinesterase inhibitor and its anti-AChE activity in the snail Lymnaea acuminata was very high in comparison to any synthetic organic pesticides (Singh & Agarwal, 1984a). In another study, Singh & Agarwal (1984b) also observed that the latex of Euphorbia royleana reduced the level of 5-hydrodxy-tryptamine (5-HT) and dopamine in the nervous tissues of Lymnaea acuminata. Singh and Agarwal (1992 a) reported that, the latices of several euphorbious plants significantly reduced the alkaline and acid phosphatase activity in nerve tissue of Lymnaea acuminata. Cheng (1971) and Amin (1972) have recorded the molluscicidal properties of Thea olesosa, Croton tiglium, Sehima argenta and Jatropha spp. Pharmacological action of Croton tiglium is due to the presence of alkaloids (Rizk, 1987). The alkaloids are naturally occurring organic bases which contain at least one nitrogen atom either in the heterocyclic ring or linked to an aliphatic skeleton. They are found in vascular plants and rarely occur in gymnosperms cryptogams and monocotyledons. Okunji and Iwu (1988), screened several plants of different families for molluscicidal properties and suggested that the toxic properties of these plants may be due to the presence of alkaloids. Toxicity in Codiaeum spp. is due to the presence of tanin in the latex (Wealth of India 1985). Tanin are complex phenolic compounds, divided in to two groups (i) The hydrolysable tanins, which one esters of gallic acid and also glycosides of these esters and (ii) The condensed tanins, which are polymers derived from various flavonoids. The molluscicidal activity was found to be related to the free phenolic groups of the tanins.
Euphorbiales as Piscicides (Table 2) The method is simple, the poisonous ingredients are pounded and thrown in to a pool or dammed up sections of a small river. After a short time the fish begin to rise to the surface and can then readily be taken by hand. The fish can be eaten without health problems (Singh, 2001). According to Neuwinger (1994),258 fish poisonous plants are present in Africa, based on 25 years of field research by the author in tropical Africa and evaluation of herbarium notes. 10-20 percent fishing poisonous are probably still unknown. They are spread among 167 plant general and 60 families. The evaluation shows a clear dominance of the leguminose Caesalpiniaceae, Mimosaceae, Papopmaceae in the hierarchy of fish poisoning plants. It also remarkable that a great proportion is in euphorbiales.
t>.l
Table 1. A list of Euphorbious plants having molluscicidal activity
.§
;:.-
Plant
Acalypha ornata
Plant part Tested Leaf, Root
Class of active moiety
Extracts
Unknown
Methanol
Alkaloids, sterols Unknown
Water Methanol
B. ferruginea
Leaf
Sterols, saponins
Water
Cryptogonone argentea Euphorbia antisyphlitica E. lactea criststa E. neutra E. pulcherima E. royleana Jatropha gossypifolia Manihot glaziovii Schima argenta Thea olesosa Uapaca guinensis
Root Latex Latex Latex Root Latex Latex Stem bark
Unknown Unknown Unknown Unknown Unknown Unknown Jatrophane Unknown
Stem bark
Croton tiglium
Latex, stem bark Latex, stem bark Latex, stem bark Latex, stem bark
Sterols, saponins Saponins, tanins Saponins, tannis Ellagic acids Ellagic acids
Water Water Water Water Water Water Water Water Water Water Methanol
Euphorbia hirta
c
&
o·
Fruit Stem bark
Euphorbia pulcherima
Reference(s)
!::
Alchornea cordifolia Bridelia atroviridis
Codiaeum variegatum
Species
Water Water Water
B. globosus B. globosus B. glabrata, B. pfeifferi B. globosus L. acuminata L. acuminata B. globosus B. globosus L. acuminata B. globosus B. globosus B. globosus B. globosus B. glabrata, B. pfeifferi L. acuminata, 1. exustus L. cuminata, 1. exustus L. cuminata,
Adewunmi & Sofowora, 1980 Okunji & Iwu, 1988 Adewunmi & Sofowora, 1980 Okunji & Iwu, 1988 Adewunmi & Sofowora, 1980 Singh & Agarwal, 1987 Singh & Agarwal, 1988a Adewunmi & Sofowora, 1980 Adewunmi & Sofowora, 1980 Singh & Agarwal, 1984a. Adewunmi & Sofowora, 1980 Adewunmi & Sofowora, 1980 Cheng, 1971 Amin, 1972. Okunji & Iwu, 1988
'"~ ~
;::s
1t ~
'"
~
;::: !::
".'"'"~
'";::s ~
R..
~
'"'"
".~
'"
Yadav & Singh, 2001 Yadav & Singh, 2001 Singh et at., 2004
1. exustus
Water 1. exustus
L. cuminata,
Singh et al., 2004
.,. 01 C.:>
454
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Table 2. A list ofEuphorbious plants having piscicidal activity Plant
Euphorbia royleana Euphorbia tirucalli Euphorbia tirucalli Glochidion velutinum Mallotus philippensis Manihot esculenta Euphorbia royleana Jatropha gossypifolia Croton tiglium Codiaeum variegatum
Plant part Tested
Class of active moiety
Extract
Species
Reference(s)
4-deoxyphorbol 4-deoxyphorbol Unknown
Water
Channa Singh & Singh, 2000 punctatus C. punctatus Tiwari et al., 2001
Water
C. punctatus
Tiwari et al., 2001
Water
O. punctatus
Kulakkattolickal, 1989
Unknown
Water
O. punctatus
Kulakkattolickal, 1989
Unknown
Water
O. punctatus
Kulakkattolickal, 1989
Latex
Unknown
Water
Latex
Unknown
Latex
Unknown
Stem bark
Taraxerol
Stem bark Stem bark Latex Stem bark Stem bark Root
Unknown Water
Channa marulias Water Channa marulias Channa water punctatus Petroleum Channa punctatus ether
Singh & Singh, 2005 Singh & Singh, 2005 Yadav & Singh, 2002 Yadav et at., 2005
The latices of several genera of the Euphorbiaceae and in particular of different species of Euphorbia have been used extensively by fisherman in different countries as fish poison of high biological activity (Watt & Breyer - Brandijk, 1976; Novock et al., 1980). The rhizome of Euphorbia biglandulosa are pounded in order to release the latex and then thrown in stagnant waters of rivers. The rapidly dissolving poison first paralyses and then kills the fish. The sap though causes in irritation in the human skin has no intoxicating effect on the people who handle the latex or eat the fish. The toxicological action of the latex can be attributed to a new class of diterpenes such as esters of phorbol, 12-deoxyphorbol, 12-deoxy12-hydroxy-phorbol, ingenal, 5-deoxyingenol, 2-deoxyingenol, resiniferotoxin and tinyatoxin (Kinghorn & Evans, 1975). It has been reported that phorbol esters interact with and activate the recently discovered protein kinase-C (Takai et al., 1977). Tadpoles were found to be most sensitive to aqueous extracts of oil cake of this plant to the presence of sterol glycoside, saponins, flavonol glycosides. A terpene, 4-deoxyphorbal the active component of plant Euphorbia tirucalli is highly poisonous to fish (Kamat & Muthe, 1995). These pesticidal compounds come from more than 50 families of angiosperms. The toxic effect of ripe fruit pulp of hingan, Balanites roxyburghii, containing saponin on fishes Glossogobius giurius, Chanda
Euphorbious Plants as Molluscicides and Piscicides
455
nama, Sarotherodon mossambica and Channa marulius have been evaluated in the laboratory. Channa marulius was found more resistant to the toxic effect of hingman fruit than other three fishes. The hingman fruit pulp used for eradication of weed and unwanted fishes from the culture ponds. Same result was also found in case of karanj, Pongamia pinnata seed on different fishes (G. giuris, Chanda nama, Oreochromis mossambicus) and these extracts are also effects the non-target aquatic organism (Mohapatra & Nayak, 1998; Mohapatra & Sovan, 2000).
ACTIVE COMPOUND PRESENT IN EUPHORBIOUS PLANTS Saponins Saponins are naturally occurring plant glycosides which form a soapy lather with water they consist of a sugar moiety and an aglycone unit. The compounds responsible for the molluscicidal activity were found to be triterpenoid saponins, with LC lOO values as low as 2 ppm (Hostettmann et al., 1982).
Alkaloids Several classes of alkaloids occur in certain genera of the family Euphorbiaceae and in particular Croton and Phyllanthus species. Imidazole alkaloids have been detected in only the genus Glochidion. Croton spp. contain several types of alkaloids, viz. isoquinolines (aporphines, e.g. sparsiflorine; proaporptines, e.g. crotonsparine and dihydroproaporphines, e.g crotosparinine), morphinandienones (e.g. crotonosine).
Flavonoids The family flavonoles, both as 0detected in
Euphorbiaceae is rich in flavonoids, particularly flavones and which have been identified from several genera. They occur and C-glycosides and as methyl ethers. The flavonoids were different parts of the plant other than roots.
Diterpeneoids Numerous species of Euhporbiaceae, which contain diterpenes, show molluscicidal properties. Those with known molluscicidal properties are Euphorbia royleana, E. antisyphilitica, E. lacteal cristata, E. pulcherima, Jatropha gossypifolia, E. hirta, E. neutra, Croton tiglium and Codiaeum variegatum (Singh & Agarwal, 1984a, 1984b, 1987b, 1988a, 1990b, 1991, 1992a, 1992b; Yadav & Singh, 2001, 2002). These plants have been used as fish poisons, insecticides and molluscicides.
Monoterpenoids a-phellandrene, myrcene, sabinene, a-terpinoline, borned, camphor, geranial, linalool, ascaridol and isoscaridol.
456
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Sesquiterpenes Lactones Aromadenderene, a-bergamoptene, p-elemene, a-humulene etc.
lridoids Iridoids are those monocyclic monoterpenoids which possess a lactone ring instead of having the p-menthane skeleton. The iridoid glycosides, ligstroside and oleuropein, have been claimed to possess activity against Biomphalaria glabrata at 100 ppm at 250 ppm respectively.
Naphthaquinones Simple naphthaquinone have considerable activity whereas prenylated and dimmer naphthaquinones are comparatively inactive against snails. The prothrombogenic drug menadione has significant molluscicidal activity.
Alkenyl Phenols The anacardiac acid component, at high concentrations, LC 100 ' induced immediate distress behaviour expressed by twitching of the tentacles (Sullivan et al., 1982). With anacardic acid, the highest mortality took place only within the first 24 h of exposure as during the subsequent 48h there were rarely any deaths.
Chalcones Chalcones are phenyl styryl ketones. 2, 4-dihydroxy-3', 6'-dimethoxy-chalcone showed molluscicidal activity at 40 ppm within 6 h against Biomphlaria pfeifferi and B. sudanica.
Furanocoumarins Molluscicidal activities of five such compounds (Xanthotoxin, Bergapten, Isopimpinellin and Chalepnsin) have been reported. These compounds possess strong toxicity against snails through their practical use is limited because they are phytotoxic.
Isobutylamides Three isobutylamides (Affinin, N-isobutyl-2E-octadienamide and fagaramide), at relatively high concentrations, have been found to be toxic to snails. Compound 102 was tested against Physa occidentalis while compounds 103 and 104 were tested against Biomphalaria glabrata and found to be lethal (Marston & Hostettmann, 1985).
CONCLUSIONS Despite all the data available for plant sources, very little is known about the active principles themselves; only about 70 natural products with recognized molluscicidal activity have been isolated. When it is considered that more than 20,000 compounds were screened in order to discover
Euphorbious Plants as Molluscicides and Piscicides
457
the synthetic molluscicide Bayluscide® the immensity of the effort required in the search for natural, highly active molluscicides can be appreciated. However, the problem is somewhat simplified by the information already available from the testing of plant material, where the activity is known but the structures of the active components remain to be elucidated. There are a very large number of plants, which contain compounds lethal to target as well as non-target organism at doses, which are much below for synthetic compounds (Shall et al., 1988; Singh et al., 1996; Amusan et al., 1997; Singh & Singh, 2005; Yadav et al., 2004). Use of such products has the additional advantage that these contain biodegradable compounds, which are less likely to cause environmental contamination. We strongly feel that if the herbaceous products are used as molluscicides or piscicides they would not only control the harmful snails and weed or predatory fish populations but would also have easy availability, easy biodegradability and greater acceptance by the users.
ACKNOWLEDGEMENTS One of the authors (Ram P. Yadav) is thankful to Council of Scientific and Industrial Research, Govt. of India, New Delhi for award of the Pool Scientist Fellowship (CSIR sanction No. 13(8190-A)IPoolJ2007 dated 4-122007) for financial Assistance.
REFERENCES Abbott, RT. (1954). The marine molluscs of Grand Cayman Island. Monogr. Acad. Natur. Sci, Philadelphia. pp. II. Adewunmi, C.O. and Sofowora, E.A. (1980). Preliminary screening of some plant extracts for molluscicidal activity. Planta Med., 39: 57-65. Adlung, K.G. and Kauth, H. (1956). Prufung der molluskiziden Engenschaften ciniger insektizider wirkstofta. Zeit, fur Ang. Zool., 43: 301-305. Agarwal, RA. and Singh, D.K. (1988). Harmful gastropods and their control. Acta hydrochim. et Hydrobiol., 16: 113-138. Amin, M.A. (1972). Preliminary report on the molluscicidal properties of Habat EI-Mallok, Jatropha sp. Trans. Roy. Soc. Trop. Med. Hyg., 66: 805-806. Amusan, O.O.G., Msothi, J.D. and Makhuba, L.P. (1997). Molluscicidal activity of Urgmw epigea. Fitot., 68: 185-186. Bali, H.S., Singh, S. and Sharma, S. (1986). The distribution and ecology of vectors snails of Punjab. Indian J. Ecol., 13: 31-37. Barry, B.D. (1969). Evaluation of chemicals for control of slugs on field corn in Ohio. J. of Econ. Entomol., 62: 1277-1279. Brar, H.S. and Simwat, G.S. (1973). Control of the common slug Laevicaulis alte Ferussae (Gastropoda), with certain chemical. J. of Res. Punjab Agnc. Univ., 10: 99-10I. Cheng, T.H. (1971). Schistosomiasis in mainland China: A review of research and control programmes since 1949. Amer. J. Trop. Med. Hyg., 20: 26-53. Daxl, R (1971). The behavior ofthe molluscicidal compounds metaldehyde Isolon and Ioxynil against slugs in field conditions. Zeitschrift fur Angewandte Zoologic., 58: 203-24I. Froyed, G. (1975). Liver fluke in Great Britain: A survey of affected livers. Vete. Record, 97: 492-495.
458
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Godan, D. (1983). Pest slugs and snails, Biology and control (Ed. Dora Godan) translated by Sheila gruber. Springer- Verlog, Berlin Heidelberg, New York. Hostettmann, K., Kizu, H. and Tomimori, T. (1982). Molluscicidal properties of various saponins. Planta Med., 44: 33-35. Hyman, L.H. (1970). The invertebrate. Vol. 6. Mollusca 1. Mc Graw Hill, New York. Jobin, W.R. (1973). Environmental control of biharziasis snail in small reservoir. J. of the Irri. And Drain. Div., 99: 365-373. Judge, F.D. (1969). Preliminary screening of condidate molluscicides. J. of Econ. Entomol., 62: 1393-1397. Kamat, D.V. and Muthe, P.T. (1995). Poisonous effect of Euphorbia tircalli on fish. J. of Ani. Morphol. and Physiol., 42(1-2): 65-68. Khajuria, J.K. and Bali, H.S. (1988). Evaluation of three molluscicides against Lymnaea auricularia. Ver. Ruf In Lab., 17: 2. Kinghorn, A.D. and Evans, F.J. (1975). A biological screen of selected species of the genus Euphorbia for skin irritant effects. Planta. Med., 28: 325-335. Kulakkattolickal, A.T. (1989). Piscicidal plants of Nepal: ripe fruit of Catunaregam spinosa (Thunb.) (Rubiaceae) and leaves of Polygonum hydropiper L. (Polygonaceae) as fish poisons. Aqua, 78: 293-30l. Marston, A. and Hostettmann, K. (1985). Plant molluscicide. Phytochem., 24: 639-652. Marston, A. and Hostettmann, K. (1985). Antifungal, molluscicidal and cytotoxic compounds from plants used in traditional medicine. Biologically active natural products (Ed. Hostettmann, K. & Lea, P.J.) Clarendon Press, Oxford, 65-88. Matsumura, F. (1985). Toxicology of Insecticides, 2nd Edn., Plenum Press; New York. Mohapatra, B.C. and Nayak, G.B. (1998). Assessment of toxicity of ripe fruit pulp of Hingan, Balanites roxburghii, on different fishes. J. Aqua., 6: 19-2l. Mohapatra, B.C. and Sovan Sahu, (2000). Toxicity of Karanj, Pongamia pinnata seed on different fishes. The 5 th Indian Fisheries Forum. 17-20 January 2000. CIFA Bhubneshwar. Narahashi, T. (1983). Pesticide chemistry, Human welfare and the environment (Miyamoto J. & Kearney, P.C. eds.) Pergamon Press; New York. Nayak, A.K., Raha, R. and Das, A.K. (1995). Organochlorine pesticides in middle stream of the Ganga river, India. Bull. Environ. Contam. & Toxicol., 54: 68-76. Neuwinger, H.D. (1994). Fish poisoning plant in Africa. Botanica Acta, 107(4): 263-270. Novock, E. Crea, A.E.G. and Falsone, G. (1980), Inhibition of mitochondrial oxidative phosphoryaltion by 4-deoxyphorbol tiresters, a poisonous constituent oflatex of Euphorbw biglandulosa. Toxicon, 18: 165-174. Okunji, C.O. and Iwu, M.M. (1988). Control of schistosomiasis using Nigerian medicinal plants as molluscicides. Int. J. Crude Res., 28: 246-252. Oteifa, B.A., Mouse, A.H., Abou-EL-haan, A.A., Mohamed, A.M. and EL-Eman, M.A. (1975). Effects of certain insecticides in the control of fresh water snails, Biomphalaria alexandrina and Brulnius trunctatus. Egy. J. of Bilha., 2: 221-243. Park, D., Minor, M.D. and Propper, C.R. (2004). Toxic response of endosulfan to breeding and non-breeding female mosquito fish. J. Environ. Biol., 25: 119-124. Rayner, J.M. (1975). Experiments on the control of slugs in potatoes by means ofmolluscicidal baits. Pl. Path., 24: 167-171. Ritchie, L.S. (1973). Chemical control of snails. In: "Epidemiology and control of schistosomiasis" (ed. Ansari N. ), Based. S. Karger, 458-532. Rizk, M.A. (1987). The chemical constituents and economic plants of the euphorbiaceae. Bot. J. of the Linean Soci., 94: 293-326. Saravanan, T.S., Mohamed, M.A., Chandrasekar, R. and Sundramoorthy, M. (2003). Freshwater fishes as indicators of Kaveri River pollution. J. Environ. Bio!., 24: 381-389. Schall, V.T., Vasconcellos, M.C., Souza, C.P. and Baptista, D.F. (1998). The molluscicidal activity of crown of christ (Euphorbia splendens var, hisloppi) latex on snails acting as intermeadiate hosts of Schstosoma mansoni and Schistosoma haemotobium. Am. J. Trop. Med. Hyg., 58: 7-10. Selvarani, D. and Rajamanickam, C. (2003). Toxicity of PCB 1232 on mitochondria of fish Arius caelatus (Valenciennes). Ind. J. Exp. Biol., 41: 336-340.
Euphorbious Plants as Molluscicides and Piscicides
459
Singh, A. and Agarwal, R.A. (1988), Possibility of using latex of euphorbiales for snail control. The Sci. of the Total Environ., 77: 231-236. Singh, A. and Agarwal, R.A. (1990). Molluscicidal properties of synthetic pyrethroids. J. of Med. & Appl. Malacol., 2: 141-144. Singh, A. and Agarwal, R.A. (1992a). Toxicity of the latex of euphorbiales. Effect on acid and alkaline phosphatase of the snail Lymnaea acuminata. Bio!. Agric. & Hortic., 8: 211-219. Singh, A. and Agarwal, R.A. (1992b). Molluscicidal activity of euphorbiales against the snails Indoplanorbts exustus. Acta. Hydrochim. Hydrobiol., 20: 262-264. Singh, A., Singh, D.K., Mishra, T.N. and Agarwal, R.A. (1996a). Molluscicides of plant origin. Bio!. Agric. & Hortic., 13: 205-252. Singh, D.K. and Agarwal, R.A. (1982). In vitro inhibition of acetylcholinesterase by carbamate and organophosphorus pesticide in the snail Lymnaea acuminata. Compo Physiol. and Ecol., 7: 191-196. Singh, D.K. and Agarwal, R.A. (1983). In VIVO and in vitro studies on synergism with Anticholinesterase pesticides in the snail Lymnaea acuminata. Arch. of Environ. Contam. and Toxicol., 12: 483-487. Singh, D.K. and Agarwal, R.A. (1984a). Correlation of the anti-cholinesterase and molluscicidal activity of the latex of Euphorbia royleana on the snail Lymnaea acummata. J. Nat. Prod., 47: 702-705. Singh, D.K. and Agarwal, R.A. (1984b). Alteration of biogenic level in the snail Lymnaea acummata by latex of Euphorbia royleana. Toxicol. Lett., 21: 309-314. Singh, D.K. and Agarwal, R.A. (1987). Latex of Euphorbw antisyphlittca, a new potent molluscicides having anticholinesterase activity against the snail Lymnaea acuminata. The Sci. of Total Envtron., 61: 211-215. Singh, D.K. and Agarwal, R.A. (1991). Action sites of cypermethrin, a synthetic pyrethroid in the snail Lymnaea acuminata. Acta Hydrochim. et Hydrobiol., 19: 425-430. Singh, D. and Singh, A. (2000). The acute toxicity of plant origin pesticides into the freshwater fish Channa punctatus. Acta hydrochim. Hydrobiol., 28 (2): 92-94. Singh, D. (2001). Studies on toxicological and biochemical effects of phytopesticides on non-target freshwater fish Channa punctatus. Ph.D. thesis DDU Gorakhpur University, Gorakhpur U.P India. Singh, D. and Singh, A. (2005). The toxicity of four native Indian plants: Effect on AChE and acid/alkaline phosphatase level in fish Channa marulias. Chem., 60: 135-140. Singh, K., Singh, A. and Singh, D.K. (1998). The use of piperonyl butoxide and MGK-264 to improved the efficacy of plant derived molluscicides. Pest. Sci., 54: 145-149. Singh, O. and Agarwal, R.A. (1981). Toxicity of certain pesticides to two economic species of snails in Northern India. J. of Econ. Entomol., 74: 568-571. Singh, S.K. and Agarwal, R.A. (1981). Effect of eyestalk ablation on oviposition in the snail Lymnaea acuminate. The Veltger., 30(3): 291-294. Singh, S.K., Yadav, R.P., Singh, D. and Singh, A. (2004). Toxic effect of two common euphorbiales lattices on the freshwater snail Lymnaea acuminata. EnVIron. Toxtc. & Pharmacol., 15: 87-93. Smith, F.F. and Boswell, A.L. (1970). New baits and attractants for slugs. J. of Econ. Entomol., 63: 1919-1922. Srivastava, V.K. and Singh, A. (2001). Possibility of using snails as bioindicators of pesticidal pollution. Iberus., 19: 1-5. Stringer, A. (1946). A note on the action of metaldehyde on slugs. Ann. Rep p. Hort. Res. Stn. Long Ashton, 87-88. Sullivan, J.T., Richards, C.S., Lloyd, H.A. and Krishna, G. (1982). Anacardic acid: molluscicide in cashew nut shell liquid. Planta Med., 44: 175-177. Takai, Y., Kishimoto, A., Inoue, Y. and Nishizuku, Y. (1977). Studies on a cyclic-nucleotide independent protein kinase and its proenzyme in mammalian tissues i.e. purification and characterisation of an active enzyme from bovine cerebellum. J. Btal. Chem., 252: 7610-7616. Tiwari, S., Sahani, S.K. and Singh, A. (2001). Control of predatory and weed fishes through environmentally safe plant origin pesticides. In: Proc. of Int. Congo Chem. Environ. (Ed. Gargh, S.L.), pp. 170-174.
460
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Tripathi, A.M. and Agarwal, RA. (1998). Molluscicidal and anti-AChE activity of tertiary mixtures of pesticides. Arch. of Environ. Contam. Toxieol., 34: 271-274. Watt, J.M. and Breyer-Brandijk, M.G. (1976). Medicinal and poisonous plants of Southern and Eastern Africa, E & S. Livingston Ltd. Edinburg London. Wealth of India, (1985). A dictionary of Indian raw materials and industrial products. Pubs. and Information directorate, CSIR, New Delhi. pp. 201-204. Yadav, RP. (2000). Studies on molluscicidal properties of some common plants of family Euphorbiaceae and their environmental impact on non-target organisms. Ph.D. thesis D.D.U Gorakhpur University, Gorakhpur U.P. India. Yadav, RP. and Singh, A. (2001). Environmentally safe molluscicides from two common euphorbiales.lberus., 19(1): 65-73. Yadav, RP. and Singh, A. (2002). Toxic effects of latex of Croton tiglium on Lymnaea acuminata and Channa punctatus. Iberus., 20(2): 31-44. Yadav, RP. and Singh, A. (2003). Effect sub-lethal concentrations of Codiaeum uariegatum latex on freshwater target snail Lymnaea aeuminata and non-target fish Channa punetatus. Nigerian J Nat. Pod. & Medi., 7: 20-24. Yadav, RP., Singh, D., Singh, S.K. and Singh, A. (2004). Toxic effect of stem bark of Croton tiglium on metabolism of freshwater snail Lymnaea aeuminata. American Malaco. Bull., 21: 87-92. Yadav, RP., Tiwari, S. and Singh, A. (2005). Toxic effects oftaraxerol extracted from Codiaeum uariegatum stem-bark on target vector snail Lymnaea aeuminata and non-target fish. Iberus., 23(1): 1-13.
Appendix Table of Contents of Other Volumes (1 & 3 to 8) ISBN: 1-933699-51-5 VOLUME
1: POTENTIAL &
CHALLENGES
1.
Biological Activity of Peptides Derived from Marine Organisms SE-KwON KIM AND Y. DOMINIC RAVICHANDRAN (REPUBLIC OF KOREA, INDIA)
2.
Bioactive Natural Products as Anti-Staphylococcal Infections SUPAYANG PlYAWAN VORAVUTIIIKUNCHAI, BEATRICE OIAWUMI lFESAN, SASITORN CHUSRI AND JONGKON SAISING (THAILAND)
23-74
3.
Pharmacological Activities, Phytochemical Investigations and In vitro Studies of Gymnema sylvestre RBr. - A Historical Review A BAKRUDEEN Au AHMED, N. KOMALAVALLI, M. MUTHUKUMAR, J.H.F. BENJAMIN, AS. RAo, SE-KWON KIM AND M.V. RAo (INDIA, SOUTH KOREA)
75-99
4.
Andirobas of the State of Acre (Brazil): Chemical and Biological Aspects Associated with the Use and Management ANA ClAUDIA F. AMARAL, JOSE LUIZ P. FERREIRA, SILVIA L. BASSO AND JEFFERSON ROCHA DEA SILVA (BRASIL)
101-129
5.
Natural Bioresource ofthe North Himalayan Region as a Source of Promising Radiation Countermeasure Agents: Lessons from Podophyllum hexandrum RAJEsHARoRA,AS. DHAKER, J. SHARMA, M. ADHIKARI, D. GUPTA, R CHAWlA, R KUMAR, A SHARMA, RK. SHARMA, S. SULTANA, G.N. QAZI, P.S. AHUJA AND RP. TRIPATHI (INDIA)
131-156
6.
Red Ginseng as a Potential Anti-Obesity Agent JI HYUN KIM AND INSOP SHIM (SOUTH KOREA)
157-174
7.
Carotenoids in Commercially Important Crustaceans from Indian Waters SACHINDRA, N.M., BHASKAR, N. AND MAHENDRAKAR, N.S. (INDIA)
175-189
8.
Therapeutic Uses of Venoms RUM! GHOSH AND VAIDEHI THANAWALA (INDIA)
191-209
1-22
462
9.
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Potentials of Bryophytes for Biotechnological Use MARKo SABOVLJEVIC AND ANETA SABOVLJEVIC (SERBIA)
211-233
10. Review: Molecular Biology Approach to Viper Venoms
235-256
PONLAPAT ROJNUCKARIN, CHUANCHOM MUANPASITPORN, PON SINGHAMATR AND JARADPONG ARPIJUNTARANGKOON (THAILAND)
11. Bee-Pollen Therapeutical Value MARIA GRAQA R. CAMPOS, CHRISTIAN FRIGERIO AND
257-277
FRANCISCO FERREIRA (PORTUGAL)
12. Herbal Products in Healthcare: Challenges and Potential MOHAMMED MOSIHUZZAMAN AND LEIV K SYDNES
279-306
(PAKISTAN,~ORWAY)
13. Anticancer Properties of Plant-Derived Food, Phytochemicals and Plant-Expressed Recombinant Pharmaceuticals KUMAR V.L. AND KUMAR v. (INDIA)
307-321
14. Global Medicinal Plants with Anti-HIV Activities
323-331
THET THET HTAR AND GABRIELA. AKOWUAH (MALAYSIA)
15.
~ew
Fungal Metabolites, as Antifungal, Herbicides and Insecticides for Biocontrol of Agrarian Plants Pests
333-389
A. EVIDENTE (ITALY)
16. On the Potential of Some ~atural Colouring Agents: An Overview ANNIE SHIRWAIKAR, ARUN SHIRWAIKAR, RICHARD LOBO AND
391-407
KIRTI S. PRABHU (INDIA, U.A.E.)
Index
423-444 ISB~:
VOLUME
1.
3: EFFICACY,
SAFETY
&
1-933699-53-1
CLINICAL EVALUATION II
Allylsulfides as Bioactive Compounds with Chemotherapeutic and/or Chemopreventive Effects
1-33
C. SCHERER, C. JACOB, M. DICATO AND M. DIEDERICH
(GERMANY, USA) 2.
Targeting Tumor Angiogenesis for Preclinical Validation of Antiangiogenic Compounds from Medicinal Plants
35-93
Appendix - Table of Contents of Other Volumes of the Series
463
BHARATHI P. SALlMATH, THIPPESWAMY G., SHEELA M.L., JOHN T. LUCAS JR. AND STEVEN A ROSENZWEIG (INDIA, USA) 3.
4.
Effects of Hydroalcoholic Extracts of Four Chinese Hypericum species on Learning and Memory in Mice DONGMEI WANG, JIE BAI, GEORGE Q LI AND DEPO YANG (CHINA, AUSTRALIA) Chemistry and Pharmacology of Shorea robusta K. PUNDARlKAKSHUDU, MD. YASEEN KHAN AND HEMANT SAGRAWAT (INDIA)
95-104
105-124
5.
Safety, Efficacy and Preclinical Evaluation of Plant Products VADDE RAMAKRISHNA AND ORUGANTI H SETTY (INDIA)
125-148
6.
In vitro and In vivo Combined Anti-Influenza Virus Effects of a Plant Polyphenol-Rich Extract and Synthetic Antiviral Drugs JULIA SERKEDJIEVA (BULGARIA)
149-165
7.
Antihypertensive and Hypolipidemic Effects of Tuber of Apios americana Medikus in SHR KUNIHIsA IWAI, SHUSUKE KURAMOTO AND liAJIME MATSUE (JAPAN)
167-181
8.
Anti-Ulcer Effects of Aqueous Extract of Persea americana Mill (Avocado) Leaves in Rats BAMIDELE V. OWOYELE, ISMAILA K. AnEBAYO AND AYODELE O. SOLADOYE (NIGERIA)
183-188
9.
Anti-Asthmatic Medicinal Plants Ezum AC. AND AKAH P.A (NIGERIA)
189-227
10. Hepatoprotective Effects of Pimpinella anisum Seed Extract in Rats NUREDDIN CENGIZ, HANEFI OZBEK AND AYDIN HIM (TuRKEY)
229-235
11. Safety and Efficacy Study of Polyherbal Formulations "Mersina", "Limit" and "Emuil" in Experimental Animals SATEESH B. AND VEERANJANEYULU A (INDIA)
237-249
12. A Review of Biological Effects of Berberine and Croatian Barberry (Berberis croatica Horvat) as a New Source of Berberine IVAN KOSALEC ,MARIJANA ZOVKO, DARIO KREMER, NADA ORSOLIC AND KsENIJA KARLOVIC (REPUBLIC OF CROATIA)
251-264
464
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
13. An Ethnopharmacological Review on Calotropis species LL. KOTHARI, M.H. PARABIA, J.H. PATEL, FARZIN M. PARABIA, SMITA S. PATHAK, FALGUNI SHETH, HIREN V. DOSHI AND M.A PATEL (INDIA)
265-285
14. Antisickling Activity and Thermodegradation of an Anthocyanin Fraction from Ocimum basilicum L. (Lamiaceae) P.T. MPIANA, V. MUDOGO, D.S.T. TSHIBANGU, KN. NGBOLUA, P.K MANGWALA, E.K ATIBu, M.K KAKuLE, L.K MAKELELE AND M.T. BOKOTA (RD CONGO)
287-295
15. Pilot Clinical Trial ofKJJ® in Children with Uncomplicated Respiratory Tract Infections S. BABAYAN, G. AsLANYAN, E. AMROYAN, E. GABRIELYAN AND A PANOSSIAN (ARMENIA, SWEDEN)
297-307
16. Antidiabetic Effect of a Herbal Powder Consisting of Curcuma longa, Emblica officinalis, Gymnema sylvestre and Tinospora cordifolia ANDALLUB., SUBHA V., VINAyKuMARAV. AND RAMA KUMAR KS. (USA, INDIA)
309-318
17. Effect of Aegle marmelos, Gymnema sylvester and Momordica charantia on Blood Glucose and Lipid Profile in Alloxan Induced Diabetic Rabbits G.B. JADHAV AND S.A KATTI (INDIA)
319-328
18. Chemical Constituents from Anti-inflammatory Fraction of Taraxacum mongolicum and Their Inhibition of AChE KEXIN HUANG, SHU YUN SHI, FENG LI, LIYAN SONG, MING YIN, ZEJIAN WANG, RONGMIN Yu, YIHANG Wu, QIAOFENG TAO, Yu ZHAO, YAN Xu, XrUMEI Wu, XIAOKUN LI, JOACHIM STOCKIGT AND J IA Qu (CHINA)
329-342
19. Inhibition of Hyaluronidase by Essential Oils and Other Natural Fragrant Extracts EMILIE ULRICH AND BENOiT AUFFRAY (FRANCE)
343-349
20. Chemical, Biological and Pharmacological Studies of the Essential Oil of Croton nepetaefolius Baill
351-362
JOSE HENRIQUE LEAL-CARDOSO, VANIA MARrLANDE CECCATTO, SELENE MArA DE MORAIS, ANDRELINA NORONHA COELHO-DE-SOUZA, ORIEL HERRERA BONILLA, SANDRA MARIA DIAS MORAIS,
465
Appendix - Table of Contents of Other Volumes of the Series
GLORIA ISOLINA PINTO DUARTE, SAAD LAHLOu AND PEDRO JORGE CALDAS MAGALHAEs (BRAZIL) 21. Pharmacokinetic Studies on Hepatic Uptake Mechanism of Tetrodotoxin in the Puffer Fish Takifugu rubripes TAKUYA MATSUMOTO AND YUJI NAGASHIMA (JAPAN)
363-391
22. Herbal Drugs - Applications in the Post Genomic Era ISHA DHANDE AND RUMI GHOSH (INDIA)
393-402
23. Antifungal Activity of Marine Bacterial Associates of Deep-Sea Origin Against Plant Pathogens A. VIMALA, C.C. RATH AND A. SREE (INDIA)
403-411
Index
425-437 ISBN: 1-933699-54-X
VOLUME
4:
ANTIOXIDANTS
&
NUTRACEUTICALS
1.
The Challenges of Antioxidant Therapy with Natural Products A.J. NUNEZ SELLES AND G. GARRIDO GARRIDO (CUBA)
1-35
2.
Antioxidant Activity of the Phytochemicals BRATATI DE AND ARCHANA BANERJEE (INDIA)
37-66
3.
Determination of Antioxidant Capacities of Non Alcoholic Beverages Prepared from Three Wild Fruits of Zimbabwe NHUKARUME L., CHIKWAMBI Z. AND MUCHUWETI M. (ZIMBABWE)
67-87
4.
Spontaneous Short-Term Fermentation of Garlic Potentiates its Anti-Oxidative Activities YOSHIMI NIWANO, EMIKO SATO AND MASAHIRO KOHNO (JAPAN)
89-98
5.
Natural Antioxidant Phytochemicals in Fruits, Berries and Vegetables and their Degradation Status During Processing SINGH DHEERAJ, BHUSHAN SASHI, LOBSANG WANGCHU, KAVITAA. AND MOOND S.K. (INDIA)
99-131
6.
The Grape Fruit Flavanone, Naringin Reduces Ferric Ion Induced Oxidative Stress In vitro GANESH CHANDRA JAGETIA (INDIA)
133-155
7.
Scavenging Capacity of Allium Species STAJNER D., POPOVIC' B.M. AND STAJNER M. (SERBIA)
157-166
466
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
8.
Effect of Aquilegia vulgaris on Liver Antioxidant Status in Rats Treated with Diethylmaleate JADWIGA J ODYNIS-LIEBERT, MALGoRZATA KUJAWSKA, IRENA MATLAWSKA, WIESLAWA BYLKA AND MAREK MURIAS (POLAND)
167-177
9.
AntioxidantActivity, Medicinal Plants and Nervous Disorders: A Review SHARMA KOMAL, SHARMA DURGESH, JAIN AYUSHI, JOSHI CRETAN AND BHATNAGAR MAHEEP (INDIA)
179-215
10. Free Radical Scavenging and DNA Damage Preventing Properties of Amaranthus tricolor L. ANANYA MUKHOPADHYAY AND BRATATI DE (INDIA)
217-227
11. Free Radical Scavenging Activity ofMethanolic Bark
229-234
Extract of Limonia acidissima THET TRET HTAR AND G.A. AKOWUAH (MALAYSIA) 12. Sorghum arundinaceum, A Wild Cereal Grain with Potential: Comparison of its Antioxidant Potential with that of Domestic Cereals in the Same Family CHITINDINGU K, BENHURA M.A.N. AND MUCHUWETI M.
235-243
(ZIMBABWE)
13. Antioxidant and Micronutrient Potential of Some Marketed Mango Varieties and Mango Products AGTE V. VAISHALI AND TARWADI V. KrRTAN (INDIA)
245-257
14. Vimang: Experiences from the Antioxidant Therapy in Cuban Primary Health Care GILBERTO L. PARDO ANDREU, ALBERTO J. NUNEZ SELLES, MARrELA GUEVARA GARciA AND AuNAALVAREZ LEON (CUBA)
259-271
15. Novel Nutraceutical Properties of Stevia rebaudiana (Bertoni) Bertoni SHARMILA CHATTOPADHYAY (INDIA)
273-281
16. Oligosaccharides and Total Polyphenols Contents in Italian Common Bean (Phaseolus vulgaris L.) Landraces: A Quality Evaluation FIBIANI M. AND Lo SCALZO R. (ITALY)
283-292
17. Biological Activities and Main Compounds of Fruits PATRICIA DALLA SANTA SPADA, GUSTAVO SCOLA, JoAoA.P. HENRIQUES AND MIRIAN SALVADOR (BRAZIL)
293-313
467
Appendix - Table of Contents of Other Volumes of the Series
18. Grape Juice: Its Compounds and Health Benefits DANI CAROLINE, OLIBONI LIVIA S., HENRIQUES JoAoAP. AND SALVADOR MIRIAN (BRAZIL)
315-326
19. Nutraceutical Potential of Commonly Consumed Fruits and Vegetables VAISHALI V. AGTE AND KIRTAN V. TARWADI (INDIA)
327-349
20. Bioactive Compounds and Functional Foods of Pseudocereals TAMER H. GAMEL (EGYPI')
351-371
387-399
IINDEX
ISBN: 1-933699-55-8 VOLUME
5: IMMUNE-MODULATION & VACCINE ADJUVANTS 1-19
1.
Herbal Therapies as Immune Modulators for Anti-Cancer Therapeutics STEPHEN M. SAGAR, DANIEL M-Y. SZE AND RAIMOND KWONG (CANADA, AUSTRALIA)
2.
Neem Leaf Glycoprotein as a New Vaccine Adjuvant for Cancer Immunotherapy BARAL RATHINDRANATH, SARKAR KOUSTAV, MANUAL-GHOSH ISHITA AND BOSE ANAMIKA (INDIA)
21-45
3.
Honey Bee Products: Immunomodulation and Antitumor Activity NADA ORSOLIC, DAMIR SIROVINA, IVAN KOSALE AND IVAN BASnc (CROATIA)
47-89
4.
Mitogenic and Anticoagulant Activity of a Fucoidan Isolated from Brown and Red Seaweed LYDICE C.M. CARVALHO, HuGO AO. ROCHA, FERNANDA W. OLIVEIRA,SUELyF. CHAVANTE, SELMA M.B. JERONIMO AND EDDA L. LEITE (BRAZIL)
5.
Development of Plant Based Adjuvants for Vaccine Antigens GUPTAAMIT, KHAJuRIAA, SINGH J., TABASUM SIDIQI, SINGH S., GUPTA V.K, SUR! KA, SATTI N.K, SRINIVAS V.K, ELLA KRISHNA AND QAZI G.N. (INDIA)
105-122
6.
Immunomodulation and Vaccine Adjuvants TEENA MOHAN, BHAT A AJAZ AND RAO D.N. (INDIA)
123-162
91-104
468
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
7.
Recent Advances in the Use of Medicinal Plants as Immunostimulants NAYAK SMITAAND MENGI SUSHMA (INDIA)
163-202
8.
Balance of Pro-IAnti-Inflammatory Cytokines Release in Spleen Cells from Mice Treated with Crotalus durissus terrificus Venom GARDuNo-RAMIREZ M.L., HERNANDEZ CRUZ A., ZUCATELLI MENDONQA R. AND PETRICEVICH V.L. (MEXICO, BRAZIL)
203-218
9.
Bioactive Agents from Herbal Sources with Immunopharmacological Properties Abating Inflammation and Malignancy CHAKRAVARTY AsHIM K., MAzUMDER TAMAL AND YASMIN HADIDA (INDIA)
219-266
10. Differential Effects of Subchronic and Chronic Oral Treatments with Orbignya phalerata Mart. Mesocarp on the Inflammatory Response FLAVIAR.F. NASCIMENTO, ELIZABETH S.B. BARROQUEIRO, ANA PAULA S. AzEVEDO, MARCIA C.G. MACIEL, WANDERSON S. PEREIRA, SUSANNE C. FERREIRA, JARDEL C. FARIAS, MAYARA T. PINHEIRO, LUCILENEA. SILVA AND ROSANE N.M. GUERRA (BRAZIL)
267-281
II. A Mistletoe Lectin-Containing Preparation for Oral Use Provokes an Immune Response and Induces an Increase in the Population of Activated Natural Killer Cells WINGE 1., DALE T.M., TIrnEM J.P.P. AND PRYME LF. (NORWAY)
283-299
12. Flavonoids from Complementary and Alternative Medicine: Mechanism of Imunomodulation of Macrophages PANDIMA DEVI K., KIRUTHIGA P.V. AND KARUTHA P ANDIAN S. (INDIA)
301-319
13. Herbal Immunomodulators CHAUHAN SINGH NAGENDRA, SHARMA VIKAS AND DIXIT V.K. (INDIA)
321-355
14. Immunomodulatory Activity of Botanicals RAUL A., KHAJURIAA., SINGH S., SINGH G.D., CHANDAN B.K. AND GUPTA V.K. (INDIA)
357-383
15. The Potentials of Immunomodulatory Substances of Natural Origin in Contemporary Medical Practice
385-403
469
Appendix - Table of Contents of Other Volumes of the Series
CHUKWUEMEKA SYLVESTER NWORU AND PETER ACHUNIKE AKAH (NIGERIA) 405-415
16. Study ofImmunomodulatory Activity of Sphaeranthus indicus VIKANI K.V., LAHIRI S.K., SANTANI D.D., KAPADIA N.S. AND SHAH M.B. (INDIA)
431-441
Index
ISBN: 1-933699-56-6 VOLUME
6: EXTRACTION,
ISOLATION
&
CHARACTERIZATION
1-55
1.
Recent Insights on the Chemistry and Pharmacology of Withasteroids MARIA LEOPOLDINA VERAS, OTILIA DEUSDENIA LOIOLA PESSOA, EDILBERTO ROCHA SILVEIRA, ANA ISABEL VITORINO MAlA, RAQUEL CARVALHO MONTENEGRO, DANILO DAMASCENO ROCHA, CLAUDIA PESSOA, MANOEL ODORICO DE MORAES AND LETICIA VERAS COSTA-LOTUFO (BRAZIL)
2.
Phenolic Compounds from Plumbago zeylanica and their Cytotoxicity NGUYEN A.-T., MALONNE H., FONTAINE J., BLANCO L., VANHAELEN M., FIGYS J., ZIZI M. AND DUEZ P. (BELGIUM, FRANCE)
57-69
3.
Extraction, Characterization and Biological Properties of 4-0-Methyl Glucuronoxylan from Hard Wood - A Review. ALINE BARBAT, CHARLOTTE MOINE, PIERRE KRAusz AND VINCENT GLOAGUEN (FRANCE)
71-95
4.
Biologically Active Naphthaquinones from Nature VINOTHKUMAR S.P. AND GUPTA JAYANTA KUMAR (INDIA)
5.
Biological Function of Glycoproteins KWANG LIM AND KYE-TAEK LIM (SOUTH KOREA, CANADA)
97-118
119-142
6.
Chemical Constituents and Pharmacology of the N eotropical Burseraceae JUNIOR VEIGA F. VALDIR AND RUDIGER L. ANDRE (BRAZIL)
143-165
7.
Are Well-Studied Snake Venoms Well Investigated? Strategy for Isolation of New Polypeptides from Snake Venom OSIPOV A.V., TSETLIN V.I. AND UTKIN Yu.N. (RUSSIAN FEDERATION)
167-184
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
470
8.
Extraction, Isolation and Characterization of Solanesol from Nicotiana tabacum L. RAo NAGESWARA R. AND NARENDRA KUMAR TALLURI M.V. (INDIA)
185-201
9.
Cinnamon: Molecular Evidence for the Health Benefits Through Its Insulin-Like and Anti -Inflammatory Effects CAO HEPING, URBANF. JOSEPH JR. AND ANDERSON A. RICHARD (USA)
203-227
10. Flax Cyanogenic Glycosides BARTHET VERONIQUE J. AND BACALA RAy (CANADA)
229-264
11. Isolation and Preliminary Characterization of Antimicrobial Proteins and Peptides from Ctenophores and Cnidaria GRANT SUZANNE, GISONDI AMY, HORTANO WILLIAM, DEFIUPPO JOHN, AND BECK GREGORY (USA)
265-286
12. Plants of the Genus: Commiphora-their Chemistry SELVAMANI P., GUPTAJAYANTA KUMAR AND SEN DHRUBOJYOTI (INDIA)
287-322
13. Secondary Metabolites and Biological Activities of some Gentianaceae species from Serbia and Montenegro SAVIKIN K, JANKOVIC T., KRSTIC-MILOSEVIC D., MENKOVICN.AND MILOSAVLJEVIC S. (SERBIA)
323-340
14. Chitin and Chitosan: Extraction and Characterization ELSABEE MAHER Z AND ALsAGHEER F AKHRIEA (EGYPT, KUWAIT) 15. Chemical Composition of the Mango Stem Bark Extract (Mangifera indica L)
341-381
383-412
NUNEZ SELLES A.J. AND RAsTRELU LUCA (ITALY, CUBA)
Index
427-437 ISBN: 1-933699-57-4
VOLUME
1.
7: STRUCTURAL MODIFICATIONS & DRUG DEVELOPMENT
Synthetic and Clinical Status of Marine Derived Anticancer Peptides t
1-28
Appendix - Table of Contents of Other Volumes of the Series
471
DIWAN S. RAWAT, RAM SINGH, NITIN KUMAR, MUKUL SHARMA AND M.S.M. RAWAT (INDIA) 2.
Microwave-Assisted Non-Conventional Esterification of Polysaccharides SROKovA, 1. AND EBRINGEROvA, A. (SLOVAKIA)
29-43
3.
Structural Modifications of Parthenin: A Sesquiterpene Lactone of Biological Potential BHAHWAL ALI SHAH AND SUBHASH CHANDRA TANEJA (INDIA)
45-76
4.
Prospects for the Development of Polyphenolic Acetate as the Potential Drug Candidate: A Review lIANvMANTHARAo G. RAJ, AJIT KUMAR, RANJU KUMARI, SEEMA BANSAL, PRIJA PONNAN, PRABHJOT SINGH, N IVEDITA PRIYA, GARIMA GUPTA, SHVETAMBRI ARORA, ANrL S. BAGHEL, SANJAY GaEL, RUCHIKA GULATI, USHA SINGH, RASHMI TANDON, DAMAN SALUJA, BILEKERE S. DWARAKANATH, ANANT N. BHAT, TAPESH K. TYAGI, AMIT VERMA, VISHWAJEET ROHIL, AsHOK K. PRASAD, EKTA KOHu, ANJANA VIJ, SURINDER K. BANSAL, VANNAN K. VIJAYAN, SUBASH C. JAlN, RAMESH C. RASTOGI, AND VIRINDER S. PARMAR (INDIA)
77-95
5.
Mining Bioactive Conformations: A Novel Methodology for Computing Predictive 3D-QSAR Models JAYENDRA B. BHONSLE AND DONALD P. HUDDLER (USA)
97-119
6.
Synthesis and Structure-Activity Relationships of Some Taxoids as Multidrug Resistance Modulator in MDR Cancer Cells LIMING BAl, TOSHIAKI HASEGAWA, JIAO BAl, MUTUMI KITABATAKE, J INLAN WANG, J UNICHI MATUBARA, JUN-ICHI SAKAI, JUNGUI DAl, SHUJUN ZHANG, WANXIA TANG, LIYAN WANG, YUHUA BAl, MINJIE LI, KATUTOSHI HIROSE AND MASAYOSHI ANno (JAPAN, PEOPLE'S REPUBUC OF CHINA)
121-155
7.
Antioxidant and N europrotective Effects of Synthetic Curcumin Analogues and Natural Phenolics Yu ZHAO, LEIXIANG YANG, KEXIN HUANG, LIHONG Hu, XIUMEI Wu, XrAoYU WANG, XrAOKUN LI, XIAOJIANG HAo, JOACHIM STOCKIGT AND JIA Qu (CHINA)
157-181
8.
Action of Plant Proteinase Inhibitors Using Biological Models MARIA LUIZA V. OLIVA AND MISAKO SAMPAlO (BRAZIL)
183-192
472
9.
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Plant Hormone Conjugates
193-245
ANDRZEJ BAJGUZ AND ALICJA PIOTROWSKA (POLAND)
10. Biological Prospective of Pinito I and its Structurally Modified Products
247-266
BHAHWAL ALI SHAH AND SUBHASH CHANDRA T ANEJA (INDIA)
11. Drug Sensitivity of Curcumin Analogues and Bioconjugates
267-298
ARCHANA PANDEY, VISHNU DWIVEDI AND KRISHNA MISRA (INDIA)
12. Structural Based Drug Design of Estrogen Receptor Beta Selective Ligands
299-314
B. BALAJI, C. SABARINATH AND M. RAMANATHAN (INDIA)
Index
329-336
ISBN: 1-933699-58-2 VOLUME
1.
8:
QUALITY CONTROL
& STANDARDIZATION
Chromatographic Techniques Applied to the Natural Products Chemistry
1-40
MARIAAPARECIDAM. MACIEL, VALDIR F. VEIGA JR., ANGELO DA CUNHA PINTO, FABIANO E.S. GOMES AND TEREZA N.C. DANTAS (BRAZIL)
2.
Quality Control of Natural Medicines by Immunological Assay System
41-56
WARAPORN PuTALUN, OSAMU MORINAGA, HIROYUKI TANAKA AND YUKlHIRO SHOYAMA (THAILAND, JAPAN)
3.
Comparison of HPLC and HPTLC Methods for the Determination of Rosmarinic Acid from Orthosiphon stamineus
57-65
GABRIEL AKYIREM AKOWUAH AND ISMAIL ZHARI (MALAYSIA)
4.
Quality Control and Standardization of Medicinal and Aromatic Plants ANJAN BHATTACHARYYA, RAJLAXMI POI, MD. AND
5.
67-87
WASIM AKTAR
N. SANYAL (INDIA)
Development of a Thin-Layer Chromatography-
89-96
Appendix - Table of Contents of Other Volumes of the Series
473
Densitometric Method for the Quantification of Podophyllotoxin in Podophyllum hexandrum ARCHANA PESHIN RAINA, S.K P AREEK AND KS. N EGI (INDIA)
6.
Standardisation of Polyherbal Formulations Using Marker Compounds
97-120
B.D. GUPTA, N.K SA'ITI, V.K GUPTA, PRABHU DU'IT AND KA. SURI (INDIA)
7.
Characteristics Variation of Lavender Oil Produced by Different Hydrodistillation Techniques
121-135
G. D. KIRAN BABU AND BIIrnAM SINGH (INDIA)
8.
Quality Control Methods for Herbal Medicines
137-179
GEORGE Q. LI, VALENTINA RAzMOVSKI-NAUMOVSKI, ESHAIFOL OMAR, AIK WEI TEOH, BENJAMIN KIMBLE, VINCENT L. QIAO, MIN-KYONG SONG, W ANNIT TONGKAO-ON, SRINIVAS NAMMI, SUILIN Mo, NANTIGA VIRGONA AND KONG M. LI (AUSTRALIA, CHINA)
9.
Evaluation of Biological Activity in Quality Control of Herbal Medicines
181-215
GEORGE Q. LI, VALENTINA RAzMOVSKI-NAUMOVSKI, ESHAIFOL OMAR, AIK WEI TEOH, MIN-KYONG SONG, W ANNIT TONGKAo-ON, SRINIVAS N AMMI, SUILIN Mo, NANTIGA VIRGONA AND KONG M. LI (AUSTRALIA, CHINA)
10. Chemical Analysis and Quality Control of Essential Oils
217-236
LAHLOU M. (MOROCCO)
11. Development and Validation of Ultraviolet Spectrophotometric Assay Method for the Powdered Leaves, Extracts and Formulations of Loranthus micranthus
237-249
UZOCHUKWU I.C. AND OSADEBE P.O. (NIGERIA)
12. Exploring Pharmacovigilance for Traditional Herbal Medicines
251-283
PuLOK K MUKHERJEE AND S. PONNUSANKAR (INDIA)
13. HPLC as a Tool for Qualitative and Quantitative Evaluation of Herbal Drugs and Formulations R. GoVINDARAJAN, D.P. SINGH AND A.KS. RAWAT (INDIA)
285-305
474
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
14. Consumer Protection and Regulatory Requirements for Herbal Drugs
307-321
N. SREEVIDYA AND S. MEHROTRA (INDIA)
Index
337-344
Index 1, 1,3,3-tetraethoxypropane 418 1, 1-diphenyl-2-picrylhydrazyl, DPPH % assay 109 12-deoxy-12-hydroxy-phorbol 454 12-deoxyphorbol 454 12-0-tetradecanoyl-13-acetate 102, 107 18-beta-glycyrrhetic acid 404 1-nitro-pyrene 341 2, 2' -azino-di [3-ethylbezthiazoline sulfonate 418 2, 4-dihydroxy-3' 456 2-aminofluorene 367 2-deoxyingenol 454 3, 4-dichlorochalcones 88 3, 4-dihydroxyphenyllactic acid 417 3, 4-epoxidation 339 3, 4-epoxidation pathway 345 3, 7-0-dimethylkaempferol 75 3, 7-0-dimethylquercetin 75 3', 4',5,6,7 ,8-hexahydroxyflavone 270 3', 4' ,5,6-tetrahydroxy-7 -methoxyflavone 270 3', 4' ,5-triihydroxy-7 -metoxy-6-Cglucopy 270 3'-demethyllariciresinol 106 3-nitrofluoranthene 341 3-0-a-arabinopyranoside 81 3-0-glucuronide 404 3-0-methylquercetin 75 4"'-0-trans-p-coumaroyl 99 4"'-0-trans-p-methoxycinnamoyl 99 4' -O-coumaroyl-isovitexine 95 4-deoxyphorbal 454 4-hydroxyderricin 54 5,7 -dimethoxy naringenin 88 5-amino salicylic acid 391 5-deoxyingenol 454
5-hydrodxy-tryptamine (5-HT) 452 5-hydroxytryptamine 144,145 5-hydroxytryptophan 241 5-methoxypsoralen 379 5-0-demethylnobiletin 86 6',7'-dihydroxybergamottin 378 6,7-dihydroxycoumarin (6,7-diHC) 335 6'-dimethoxy-cha1cone 456 6-gingerol 54 6-thioguanine 341 7-ethoxy coumarin (7-EC) 340 7-hydroxy coumarin (7-HC) 334, 340, 377 7 -hydroxylase 338 7-0-methyleridictyol 88 a amylase 22 a glucosidase 22 a methyl norepinophrine 391 aI-antitrypsin 165 a2-macroglobulin 146 a-bergamoptene 456 a-bisabolol 72 a-glucosidase inhibitors 220 a-glycosidase 108 a-humulene 456 a-lipoic acid 19, 20 a-methyldopa 391 a-phellandrene 455 a-pinene 69 f3-blockers 392 f3-cells 220, 225 f3-elemene 456 f3-naphthoflavone 339
A Abortion 282 Absorption 333
476
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Acalypha ornata 453 Acarbose 20 ACAT 359 Acetogenins 232, 294 Acetonitrile 418 Acetylation 389, 395 Acetylcholine (ACh) 260 Acetylcholinesterase (AChE) 451, 452 Acetylsalisilic acid 93 Achillea aleppica 72 A. millefolium 72 A. schischkinii 72 A. wilhelmsii 72 ACM 5,6295 Acrophobia 215 Actinomycete saccharopolyspora erythraea 4 Actinoplanes teichomyceticus 4 Activator protein (AP-1) 47 Acupuncture 181 Acute diarrhoea 212 Acute inflammation 135 Acute toxicity 18, 72, 75, 220, 241, 267 Ad libitum 214, 223 Adaptogens 60 Adenine 123 Adenine arabinoside 2 Adhesion molecule 166 Adiantum tetraphyllum 212 Adipocytes 442 Adrenaline 397 Aegerolysins 252, 253 Aerosol sprays 390 Aethusa cynapium 211 Affinin 456 Agrocybe aegerita 252 Ajugol99 Alanine aminotransferase 407 Alchornea cordifolia 453 Aldicarb 451 Aliphatic hydroxylation 393 Alkaloids 231, 449 Alkenyl phenols 449 Alkylating drugs 122 Allergy 39 Allopathic medicine 68 Allopurinol 397 Alloxan 222 Alteplase 410 Alternative medicine 180
Amazonial ethnomedicine 122 Amitriptyline 241, 242, 246, 391 Amitriptyline hydrochloride 241 Amorphophallus konjac 20 Amoxicillin 408 Amphibious 450 Ampicillin 269, 391 Amycolatopsis orientalis 4 A. rifamycinica 4 Anacardiac acid 456 Anacardiaceae 68 Analgesic 4, 34 Anaphylactic shock 141 Anaphylatoxins 153 Ancillary analyses 183 Andrographis paniculata 419 Angelica sinensis 53, 54 Angiogenesis 32, 46, 150 Angiogenin (AG) 47 Angiopoietin (APN) 46, 47 Angiotropin (AT) 47 Ankylosis 108 Annofolin 295 Annona cornifolia 231, 293 Annonaceae 231, 293 ANOVA 214,242,276,298,434 Anthelmintic 70 Anthocyanoids 77 Antiallergic 402 Anti-alzheimer's 4 Antiangiogenic 46 Antiarrhythmic 310, 392 Anti-atherogenic 359 Antibacterial 34, 293, 355, 418 Antibiotics 4, 5, 33, 407 Anticancer 5, 46, 79 Anticarcinogenic 122, 128 Anticlastogenic 128 Anticoagulant 5 Anticoagulation 47 Anticonvulsant drugs 337 Anticonvulsants 392 Antidepressant 239, 392 Antidiabetic 5, 219 Antidiarrhoea 231 Antidysentery 231 Antiedema 231 Antiexudative effect 381 Antifertility 5 Antifever 231
Index Anti-flatulence 70 Antifungal 235 Antigen-antibody reactions 138 Antigenotoxic 122 Anti-helicobacter pylori activity 268 Anti-hepatotoxic 79 Antihypertensive 5 Anti-inflammatory 67, 122, 219, 355, 356,402,418 Anti-influenza 231 Anti-insomnia 231 Antileukemic 2, 34 Antimalarial 5, 293, 294 Antimicrobial 34, 72, 122, 219, 231, 294 Antimutagen 418 Antimutagenic 122, 128, 367 Antineoplastic 343 Antinociceptive 67 Anti-osteoporotic 79 Antioxidant 122, 265, 355, 402, 417 A. actinomycetemcomitans 376 Antioxidation 374 Antioxidative 418 Antiparasitic 294 Anti-peroxidative activity 80 Antiperspirant 333 Antiplatelet 5, 410 Antiradicals 367 Antirheumatism 295 Antisecretory 265 Anti-sindbis activity 376 Anti-spasmodic 5 Antistressor 310 Antithrombin 400, 402 Antitumor 231 Antitussive 69 Anti-ulcer 266, 365 Antiulcerogenic 266, 267 Antivirus 34, 402, 418 Anxiety 144, 211, 245 Aphrodisiac 69 Aphtha 34 Apigenin 121, 270, 360 Apocynum venetum 212 Apolipoprotein B 360 Apoptosis 342, 370 Aporphine 73 Appetizer 69 Apratoxins 6 Aquaculture 449
477 Arachidonic acid 77, 142, 147, 275 Arceuthobium oxycedri 90 Arglabin 5 Aromadenderene 456 Aromatic acids 213 Aromatic herbs 70 Aromatic hydroxylation 393 Aromoline 73 Arrhythmias 189 Artemia salina 233, 295 Artemisia abrotanum 211, 216 A. annua 54 A. glabella 5 Artemisinin 54, 294 Arterial blood pressure 259 Arterial hypotension 313 Arthritis 441 Ascaridol 455 Ascorbic acid 121, 122, 126, 212 Aspartate aminotransferase 342, 407 Aspergillus fumigatus 49, 252 Asp-hemolysin 252, 258 Aspirin 2, 72, 365, 410 Asthma 69, 75 Astragalin 108 Astringent 418 Aterospermidine 295 A-terpinoline 455 Atherosclerosis 18, 87 Atropa belladonna 5 Atropine 5, 259, 267,269 AUC 400 Aucubin 99 Auriculasin 93 Aurilides 6 Auscultation 32 Autoimmune 97, 142, 159 Autoimmunity 441 Avena sativa 20 Ayurveda 3 Ayurvedic system of medicine 219 Aztreonam 4
B Bacampicillin 391 Bacillus polymyxa 4 B. subtilis 4, 296 Bacitracin 4 Bacteriostatic activity 235 Baicalein 54, 56, 367
478
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Balanites roxyburghii 454 Baphicacanthus cusia 33 Barbiturates 392, 395 Basic fibroblast growth factor (BFGF) 47 Basophils 140 Benzo[a]pyrene 367 Benzodiazepines 211 Berbamine 73 Berberine 73 Berberis integerrima 74 B. oblonga 73 B. paraspecta 53 Bergapten 379, 456 Beta-carotene 372 Beta-carotene-linoleic acid 375 Biexponential 403 Bifidobacterium sp. 407 Biguanidines 220 Bilberry 333 Bioactive compounds 67 Bioactivity 1 Bioassay-guided processing 67 Bioavailability 307, 334, 356, 405, 425 Biochanin A 360 Biochemical changes 32 Biodegradable 451 Bioflavonoids 359, 363, 367, Biological evaluation 1 Biomarker 9 Biomedical theory of cancer 50 Biomphalaria glabrata 456 B. pfeifferi 456 Biosynthesis 1, 157, 359, 451 Biotechnology 261 Biotransformation 307,310, 389, 405 Bisbenzylisoquinoline 73 Bivalirudin 5 Bivalves 450 Bleomycin 5, 372 Blood deficiency dryness 32 Blood plasma 320 Blood stasis 32 Borned 455 Botanicals 122 Bovine aortic endothelial cells (BAEC) 50 Boyden chamber 51 Bradycardia 259 Bradykinin 70, 141, 145 Branchiodialator 122
Bridelia atroviridis 453 Brine shrimp 234, 304 Bromodeoxy uridine 124 Bronchitis 401 Brucellosis 333 ~-sitosterol 295 Bupleurum chinense 56 Butylated hydroxytoluene 425
c C NMR spectra 298 Caecal microflora 336 Caesappiniaceae 452 Cafeoyl quinic derivatives 271 Caffeic acid 417,426 Caffeine 432 Call una vulgaris 78 Camellia sinensis 54, 194, 430 Camphor 455 Camptothecin 16 Cancer 18, 294 Candida albicans 233, 234, 236 Capillary electrophoresis 308 Carbamate 451 Carbamazepine 337, 393 Carbuncles 34 Carcinogenicity 331,342 Carcinoma 343, 367 Cardiac troponin 362 Cardiomyocytes 261 Cardioprotective 79 Cardiorespiratory effect 251 Cardiotonic 5, 259 Cardiovascular 258, 313, 355, 430 Carminative 70 Carnivorous 449 Carrageenan 141 Carrageenan-induced hind paw oedema 70 Carrageenan-induced paw edema 430 Caspases 156, 157 Catalase 360 Catalpol99 Catalposide 103 Catalytic 440 Catharanthus roseus 53 Caveolin 257 Cchrysin 378 cDNA synthesis 439 Cell-cell interactions 253
Index Centella asiatica 212 Cephalosporin 4 Cephalosporium acremonium 4 C-glucopyranoside 266 C-glycosyl flavone 79, 95 Chalcones 381, 456 Chalepnsin 456 Chanda nama 454 C. marulias 454 C. punctatus 454 Chemical mediators 68 Chemoattractants 142, 152 Chemopreventives 23, 102 Chemotactic factors 154 Chemotherapeutic 356 Chemotherapy 121, 129 Chick embryo chorioallantoic membrane (CAM) model 50 Chicory 333 Chlamydia psittaci 283 C. abortus 282 C. pecorum 282 C. psittaci 282 Chlamydiosis in dairy cattle 281 Chloramphenicol 4,234,297,391 Chloroquine 302 Chlorpropamid 19 Cholagogue 78 Cholangiocarcinoma 342 Cholera toxin B-subunit 257 Choleretic phenolic acids 212 Cholesterol 252, 390 Cholesterol-HDL 360 Cholesterol-LDL 360 Cholesterol-VLDL 360 Cholinergic effects 451 Chondrocytes 254 Chondrodendron tomentosum 5 Chromatograms 271, 423 Chromium 19 Chromobacterium violaceum 4 Chromosomal aberrations 121, 125, 370 Chronic inflammation 135 Chronic intestinal catarrh 92 Cimetidine 267,268,274, 408 Cimicifuga racemosa 211 Cinchona bark 5 Cirrhosis 337, 407 Cistus laurifolius 75 C. aurantium 212
479
C. paradisi 376 C. unshiu 381 Clarithromycin 269, 392 Clematis 95 C. cirrhosa 95 C. erecta 211 C. flammula 95 C. mandshurica 82 C. vitalba 95 Clinical efficacy 33 Clinical evaluation 1 Clinical interventions 179 Clinical studies 158, 343 Clinical trials 52, 179 Clopidogrel 410 Clorazepate 201 Clostridium butyricum 407 Cloudberry 333 CLtota1400 Cmax 406 C-NMR 269 CNS depressant activity 71 Cocaine 5 Codeine 391, 392 Codiaeum spp 452 C. variegatum 453,454 Colchicine 123 Colistin 4 Collagen matrix 363 Collagen-induced arthritis 430 Columbamine 73 Column chromatography 78, 232 Combinatorial compound library 2 Combretastatin 16 Comet assay 371 Complement system 153 Complementary 180 Conjugation 389 Conjunctivitis 282 Consort 180 Convulsions 212, 224 Coptis chinensis 53 Coriandrum sativum 212 Cornea implantation 50 Coronary heart disease 191 Corpus luteum 122 Corticosteroid 401, 410 COSY spectrum 298 Coumarins 77, 331, 400 COX-1 74, 133,148
480
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
COX-2 74, 133, 148, 368, 372, 435 C-reactive 156 Croton tiglium 452, 453 Cryptogonone argentea 453 Cumene hydroperoxide 373 Curcuma longa 54 Curcumin 54, 368 Curcuminoids 57 Cyamopsis tetragonoloba 20 Cyanobacteria 6 Cyanosis 258 Cyclization 389 Cyclooxygenase (COX) and lipoxygenase (LOX) 47 Cyclooxygenase 147, 174 Cyclosporine 2, 32 Cyclosprin 5 Cynomolgus monkey 343 CYP1A1I2 393 CYP2A6337 CYP2C19393 CYP2C8/9 392 CYP2D6392 CYP2E1393 CYP3A 4/5 392 CYP3A4 activity 379 CYP-dependent pathways 338 Cytokines 32, 134, 150 Cytolytic 252 Cytoplasmic vacuolation 342 Cytoprotective effect 365 Cytosine 2 Cytosine arabinoside 2 Cytoskeletal 369 Cytosol 390 Cytostatic 59, 102, 294 Cytotoxic chemotherapy 45 Cytotoxicity 165, 234
D D-400 221 Daphne acutiloba 106 D. genkwa 106 D. oleoides 106 D. tangutica 106 Daptomycin 4 Daunorubicin 5 DBAl2338 Deamination 393 Decamethonium 390
Decyclization 389 Delirium 212 Demulcent 401 Depression 144, 308 Depurative 78 Dereplication 1 Dermatitis 39 Dermatoses 34 Desacetyldiltiazem 379 Desmethyl 391 Destagnation herbs 53 Detoxicating 34 Detoxification pathway 343 Dexamethasone 339 Dexfenfluramine 246 Dextran 144 d-glucose 223 Diabetes 313, 430 Diabetes mellitus 219 Diadzein 364 Diaphoretic 78, 107 Diarrhoea 287 Diazepam 80, 215, 241, 391, Dicumarol410 Dietary supplements 19 Digitoxin 391 Digoxin 4, 391, 410, 412, Dihydrobinetin 368 Dihydrocoumarin (DHC) 335 Diltiazem 379, 392 Dimers 254 Dimethylaminoethanol bitarate 203 Dimethylformamide (DMF) 296 Dimethylsulfoxide 233 Dimeton 451 Dioscorea deltoideae 5 Diosgenin 5 Dipyridamole 410 Dislipidaemia 18 Distribution 333 Diterpeneoids 455 Diterpenes 454 Diuretics 69, 72, 78, 201, 414 DMSO 233 DNA cleavage 367 DNA replication 123 DNA synthesis 341 Dopamine 391, 396, 452
481
Index Doxorubicin 5 DPP-IV 22 Dragendorffs reagent 233 Dropsy 98 Drug discovery 2 Drug interactions 355, 356 Drug metabolism 332 Drug-metabolizing enzymes 390 Drynaria fortunei 363 d-tubocurarine 4, 5 Duncan's multiple range test 434 Dunkin-hartley guinea pig 343 Dunnett's test 269, 276 Dysmenorrhea 213 Dyspepsia 97, 213
E Ebers papyrus 3 Ecchymoses 34 Echinacea angustifolia 195 E. pallida 195 E. purpura 195 Eczema 34, 71, 75, 92 Edema 47 Efficacy 1, 180 EGCG 431 Eicosanoid release 88 Electrocardiograms (ECG) 258 Electropherogram 315 Electro-retinography 360 Eleutherococcus senticosis 194 Elevated plus-maze model 215 ELISA 11 Emmenagogue 70 Enalapril 391 Enalaprilat 391 Encephalomyelitis 282 Endocrine features 32 Endocrine system 356 Endocytosis 253 Endogenous 390 Endogenous mediators 142 Endogenous polypeptides 46 Endoplasmic reticulum 390 Endotoxin 142, 371, 372 Enterocytes 225 Enterohepatic 400, 402, 403 Environmental effects 32 Environmental pollution 449
Enzyme inhibitor 122 Enzyme leakage 189 Eosinophils 140 Ephedrine 4 Epibatidine 4 Epicatechin 432 Epicatechin gallate (ECG) 432 Epidemiological 20 Epigallocatechin gallate 364 Epigallocatechin (EGC) 54, 432 Epigallocatechin gallate 431 Epigallocatechin-3-gallate (EGCG) 432 Epilepsy 34, 88 Epimedium sp. 74 Epinephrine 396 Epipedobates tricolor 4 Erica bocquetti 77 E. manipuliflora 77 Eriodictyol 88 Eruption 34 Eryngium billardieri 70 E. campestre 69 E. dauisii 70 E. falcatum 70 E. isairicum 70 E. kotschyi 70 E. maritimum 69 E. trisectum 70 Erysipelas 34 Erythema 39 Erythematous papules 32 Erythrocytes 255, 314 Erythrodema 39 Erythroid 153 Erythromycin 4, 392 Erythroxylum coca 5 Escherichia coli 122, 269, 296, 376 ESI mass spectra 301 Essential oils 77, 212 Estrogens 122 Ethnobotanical 68 Ethnopharmacological 294 Ethnopharmacology 67 Etoposide 5 Eubacterium sp 407 Eugenia jambolana 219 Euhorbiales 449 Euphorbia antisyphlitica 453 E. biglandulosa 454
482
Camp. Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
E. hirta 453 E. pulcherima 453 E. royleana 452, 454 E. tirucalli 454 Euphorbiales 452 Excretion 333 Exenatide 20 Exocytosis 253 Exogenous 321 Exogenous mediators 142 Expectorant 78, 401 Exponential 400
F Factor XII 140 Fagaramide 456 Fasciola gigantica 450 F. hepatica 450 Fascioliasis 449, 450 Fatigue 308 Fatty acid methyl esters 231, 234 Fatty acids 232, 254, 295 Fatty acids fraction 234 Febrifuge 67 Felodipine 377 Ferulic acid 426 Fibroblasts 47 Fibrosarcoma 255 Ficus racemosa 22 Filaggrin 37 Fisetin 365,381 Flavoglycoside 372 Flavone glycosides 212 Flavones 265, 400 Flavonoids 75, 240, 266, 355, 360, 418, 449 Fluorescence polarization (FP) 12 Fluoxetine 246 Folk medicines 67, 221 Forced swimming test 239 Free fatty acids 431 Free radical 92, 164,431 Free radical scavenger 92, 109, 122, 128 Fucose 362 Fumagillin 49 Furanocoumarins 456 Furuncles 34 Fusidic acid 4 Fusidium coccineum 4
G GABA receptor 247 Galangin 367 Galantamine hydrobromide 4 Galanthus nivalis 4 Galega officinalis 221 Galegine 221 Gallic acid 452 Gamma-glutamyltransferase 342 Gamma-radiation 370 Gas chromatography (GC) 8 Gastric ulcer 268 Gastritis 74 Gastrointestinal 39, 334 Gastrointestinal microflora 336 Gastropods 450 Gastroprotective 265 GC-MS 69,232 Gene expression 39, 430, 439 Gene transcription 157 Genetic effects 129 Genetic polymorphism 346 Geniposidic acid 99, 100 Genistein 360, 367 Genotoxic 121, 372 Genotoxicity 332 Gentamicin 4 Gentiana arisanensis 80 G. olivieri 79 Geranial 455 Geranium 81 Geranium maculatum 81 G. pratense 81 G. sanguineum 81 G. thunbergii 81 Geriatric pharaton® 202 GericompleX® 202 Gingivitis 34 Ginkgo biloba 54, 189, 212 Ginsana® 202 Ginseng 19 Ginsenosides 197 Glimepiride 19 Glioma cells 368 Glipizide 19 Glochidion velutinum 454 Glossogobius giurius 454 Gluconeogenesis 366 Glucose transporter 430
Index
483
Glucose-6-phosphatase 342 Glucosides 333 Glucuronidation 356,357, 395 Glucuronides 357 Glucouronides conjugation 389 Glutathione 360 Glutathione conjugation 389, 395 Glyburide 19 Glycerophospholipids 257 Glycine conjugation 389, 395 Glycine max 20 Glycoalkaloids 449 Glycogenesis 220 Glycolysis 220 Glycosides 399, 452 Glycosylated flavonoids 368 Glycyrrhetic acid 399 Glycyrrhiza 399 G. glabra 400 G. uralensis 56 Glycyrrhizin 399 Gnotobiote 404 Goiter 69 Gonorrhea 213 Gram negative 233 Gram positive 233 Granulocyte-colony-stimulating factor (G-CSF) 47 Granuloma 76 Grapefruit 355 Green tea 429 GSH S-transferase 342 Guaiacam officinale 122 Guanine 123 Gymnema sylvestre 19 Gymnosperms cryptogams 452
H H NMR data 300 Haementeria officinalis 5 Haemorrhoid 72 Hageman factor 140, 142 Headaches 295 Health supplements 355 Hectochlorin 6 Helichrysum arenarium 73 H. plicatum 73 Helicobacter pylori 266 H. orientalis 96 Hematocrits 358
Hematopoiesis, 150 Hemoglobin 314, 371 Hemolysis 251 Hemolytic 252 Hemorrhoids 69, 92 Heparin 419 Hepatic 340 Hepatic necrosis 342 Hepatitis 190, 343, 399, 402, 407 Hepatitis A 337 Hepatocyte growth factor (HGF) 47 Hepatocytes 335, 339 Hepatomegaly 343 Hepatoprotective 402 Hepatotoxicity 32, 333, 342, 395 HepG2341 HepG2 cells 372 Herbal drugs 219 Herbal extract 309 Herbal interventions 180 Herbal medicines 3, 46, 179, 430 Herbal remedies 67 Hesperidin 88, 356 Heterogeneity 190 Heterogeneous 181, 365 Hexamers 254 Hexosamine 362 Hexose 362 Hexose monophosphate shunt 220 High throughput screening 2 High-fructose diet 430 High-performance liquid chromatography (HPLC) 197 Hirudin 5 Histamine 140, 143, 274, 396, 408 Histopathological 361 HMG-CoA reductase 359 HMQC spectrum 298 Hofmann elimination 397 Homeostasis 397 Homology 289 Honokio154 HPCE analyses 314 HPLC 8, 240, 357, 403, 417 HPLC-PDA method 84 HPLC-PDA system 92 HPLC-UV-PDA analysis 271 HPTLC 8 hSHBG 366 Human chondrocytes 256
484
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Human myelocytic leukemia 40 Human neutrophil functions 88 Human umbilical vein endothelial cells (HUVEC) 50 Huo Xue Hua Yu 53 HuR 438 HUVEC 55, 260 Hydrastis sp. 74 Hydrochloric acid 267 Hydrochloride 241 Hydrolysis 389 Hydroxy coumarins 212 Hydroxycinnamic 109 Hydroxycoumarin (HC) 335 Hyperforin 240 Hyperglycemia 18, 220, 361 Hypericin 196, 240 Hypericum erectum 240 H. hubeiense 240 H. perforatum 194, 240 H. seniavinii 240 Hyperkalaemia 251, 254 Hyperlipidemia 361 Hyperplasia 342 Hyperproliferation 37 Hypersensitivity 39, 41 Hypertension 83, 87 Hypocholesterolemic 359 Hypoglycaemic 219, 366 Hypokalemia 400,409, 410 Hypolaetin-8-glucoside 86 Hypolipidemic 5, 362 Hypothermic restraint stress ulcer 268 Hypoxia 259
I IL-1a 150 IL-1b 150 Ilwensisaponin A 99 Imidazole 455 Imipramine 241 Immune persistence period test 283 Immune system 149 Immunoblasts 139 Immunocompromised (ICm mice 341 Immunocytes 46 Immunogenecity 288 Immunoglobulin G 165 Immunohistochemical 37 Immunological disorders 32
Immunomodulatory 37, 333, 402 Immunosuppressive 2, 294 In vitro assays 50, 333, 377 In vivo 69,75, 294,295, 333, 377, 390, 425, In vivo assays 51 Inactivated vaccine 281 Indigo naturalis 31 Indirubin 31, 35 Indomethacin 69, 72, 158 Indoplanorbis exustus 450 Infantile cholera 212 Infantile convulsions 34 INF-b 48 Infection 32 Infertility 69, 282 Inflammation 133, 430 Inflammatory 32 Inflammatory bowel disease 430 Inseticide 231 Insomnia 78, 213 Insulin 2, 220, 431 Insulin signal transduction pathway 436 Insulin signaling pathway 430, 432 Insulin-dependent diabetes mellitus 220 Insulin-like growth factor I and II (IGF1 and II) 47 Integrins 134, 168 Interleukin 106 Interleukin-8 (11-8) 47 Interleukine-1 142 Interrogation 32 Intestinal system 356 Intestinal uptake 220 Intracellular signaling pathways 54 Involucrin 37 IR spectra 232 lridoids 104, 456 Irinotecan 16 Isatis cappadocia 75 1. glauca 75 1. indigotica 33 Ischemia 261 Islets of langerhans 225 Isoamylene guanidine 221 Isobutylamides 456 Isocoumarin 266 Isoflavones 213, 400 Isolan 451 Isolariciresinol 106
485
Index Isoorientin 79, 92 Isoorientin-6"-0-glucoside 80 Isopimpinellin 456 Isoprotereno 362, 363 Isoquinoline alkaloids 294 Isoquinolines 73, 455 Isoscaridol 455 Isovitexin 92 Itraconazole 392
J Jatropha spp 452 J. cilliata 80 J. gossypifolia 453, 454 Jatrorrhizine 73 Juglans regia 82 Juniperus oxycedri 90
K Kaempferol 3-0-~-galactopyranoside 81 Kaempferol 79, 365 Kaem pferol-3, 7 -0- a-dir hamnoside 107 Kaempferol-3-0-~-D-galactoside 78 Kallikrin 140 Kampo-medicines 407 Kanamycin 4 Karathane 451 KDRlflk-1 55 Kedde's reagent 233 Keratinocytes 31,37,38 Keratolytic 82 Ketoconazole 234, 392 Knockout mice 441
L Lagerstroemia speciosa 19, 20 Lamium album 83 Lansoprazole 267, 393 L-arginine 162 Lariciresinol 106 Larrea diuaricata 122 L. tridentata 5 Larvicidal activity 231, 293 Laurocerasus offficinalis 97 Laurus nobilis 87 Laxative 219, 414 LC-NMR 10
LDL oxidation 360 LDL receptor knockout 360 Leiothrix celiae 266 L. flauescens 265 Leucorrhoea 83 Leukemia 370 Leukocytes 133, 138, 314 Leukotrienes 68, 77, 134, 140 Levodopa 391 Lewis rats 372 Licorice 400 Limit of quantification (LOQ) 420 Limonin 368 Linalool 455 Lincomycin 4 Linoleate 234 Linoleic acid 80 Lipid peroxidation 372, 417, 424 Lipid rafts 251 Lipophilic barbiturates 390 Lipophilic drugs 389 Lipopolysaccharide 369 Lipoxygenase 79 Lipoxygenase enzyme 77 Liquid chromatography (LC) 8 Liriodenine 295 Lithium 389 Locomotor activity 71 Loganic acid 104 Lovastatin 359 LTB 140 L-theanine 432 L-tyrosine 223 Lumbago 73, 106 Luteolin 270, 276 Luteolin 0- 266 Luteolin-6-C glucoside 79 Lycopodine 91 Lycopodium clauatum 91 Lymnaea acuminata 450, 452 Lymphocytes 31, 38, 121, 129, 139 Lymphoid cells 139, 153 Lyngbyabellins 6 Lysophosphatidylcholine 252 Lysophosphatidylinositol 254 Lysophospholipids 254 Lysosomes 390 Lysyl-bradykinin 145 Lythrum salicaria 91
486
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
M Maclura pomifera 92, 93 Macrophages 76, 136, 139,275,333, 372, 435,441 Magnoflorine 73 Magnolia obovata 54 Malaria 294 Mallotus philippensis 454 Malondialdehyde 269, 424 Maltodextrin 196 Mandibular lingual region 363 Manihot esculenta 454 M. glaziovii 453 Marasmus 212 Martynoside 103 Masoprocol 5 Mass spectroscopy (MS): 9 Mastitis 282 Matrigel50 m-coumaric acid 426 MDA assay 421 Mean residence time (MRT) 409 Medicinal plants 67, 430 Melanoma cells 59 Menopausal disorders 213 Menorrhagia 83 Menstrual disorders 212 Mental fatigue 308 MeOH extract 267 Mesurol451 Metabolic activation 122 Metabolic disorders 220 Metabolic syndrome 18, 430 Metabolism 275, 331, 334, 357, 389, 402, 406, 426 Metabonomics 9 Metaldehyde 450 Metalloproteinase 56 Metanauplii 233, 296 MetAP-249 Metastases 45, 53 Metformin 20, 221 Methemoglobin 371 Methiocarb 451 Methotrexate 32 Methoxyflavone 86 Methylation 389, 395 Methyl-~-cyclodextrin (MbCD) 256 Metronidazole 408
Mevastatin 5 m-hydroxyphenylpropionic acid 426 MIC 233 MIC values 269 Microaerobic 268 Microcystin-LR 369 Microheterogeneity 257 Micrometastasis 48 Micromonospora 4 Microorganisms 7, 282, 331, 425 Microsomal 389 Microsomes 80 Miglitol 20 Migraine 144 Migraine prophylaxis 192 Mimosaceae 452 Mineralocorticoid activities 401 Miristate 234 Mithramycin 5 Mitochondria 374, 390 Mitochondrial enzymes 363 Mitomycin 5 Molecular biology 46 Molecular diversity 1 Molecular markers 51 Molecular pathways 45 Molecular targets 429 Molluscicides 449 Momordica charantia 19, 219 MOMP genes 288 Monoamine oxidase 247 Monocotyledons 452 Monocytes 139 Monomers 253 Monoterpenes 69 Monoterpenoids 455 Morin 360 Morphinandienones 455 Morphine 2, 4, 391 Mouse somatotrophs 256 mRNAs 148, 339, 431, 434 MRT 400 MS data 10 Multidrug-resistant (MDR) 368 Multiple drug targets 2 Mumps 34 Mupirocin 4 Muscarinic blocker 259 Mutagenic 123, 340 Mutagenicity 333
487
Index
Mutagens 341 Myasthenia gravis 154 Myeloid 153 Myeloid hyperplasia 441 Myocardial infarction (MI) 18, 362 Myocardial ischemia 189, 259 Myrcene 455
N NADPH 162 NADPH-oxidase 170 Narcotics 313 Nardostachys jatamansi 212 N aringenin 355 N aringin 355 Nasopharyngeal cancer 53 N ateglinide 19 Natural health products 46 Natural products 1, 2,122,220,294,331, 370,418,451 N auplii 233, 296 N -dealkylation 393 Necrosis 46, 342 Necrotizing action 274 Nei-kuan 191 Neoeriocitrin 363 N eohesperidin 368 Neomycin 4 Neoplasms 46 Neoplastic 122 Neostigmine 390 N eovascularization 46 Nepetin-7 -O-~-D-arabinopyranoside 266 N epetin-7 -O-~- D-gI ucopyranoside 266 Nephrotoxic 342 Nephrotoxicity 32, 371 Nerium oleander 71 Nervous tension 213 Nervous-mental diseases 308 Netilimicin sulfate 4 Neuralgias 295 Neuroleptics 392 Neuromuscular junction 451 Neuroses 308 Neurosis-like disorders 308 Neurotoxicants 451 Neurotoxicity 122, 450 Neutrophils 139 NF -kappa 372 NF-KB 166
NHEKs 39 n-hexane 69 Niclosamide 450 Nimodipine 377 N-isobutyl-2E-octadienamide 456 Nitric oxide 159 Nitroquinoline n-oxide 367 Nitrosothiols 162 N-methyl-N-propargyl-benzylamine 241 NMR 269 Nobiletin 367 Non-insulin dependent diabetes mellitus 220 Nonmicrosomal 389 Noradrenaline 364 Nordihydroguaiaretic acid 121, 122, 127 Norepinephrine 241,246 Nortriptyline 391 Norverapamil 379 N-oxidation 393 NSAIDs 68, 274 Nuclear factor-kappa b (NF-KB) 47 Nuclear magnetic resonance spectroscopy (NMR) 9 Nucleophilic 129 Nucleoside 2 Nutraceuticals 19 Nuvan 451
o Obesity 429 O-coumaric acid (o-CA) 335 O-dealkylation 393 Oedema 69, 136, 138, 333 O-HPAA 336 O-hydroxyphenylacetaldehyde (0- HPA) 335 O-hydroxyphenylacetic acid (o-HPAA) 335 O-hydroxyphenylethanol (o-HPE) 335 O-hydroxyphenylpropionic acid (0HPPA) 335 Oil-adjuvant vaccine 281 Okadaic acid 369 Oleate 234 Oleoresin 68 Oligomerize 253 Oly 252 Omega-3 fatty acids 20 Omeprazole 393
488
Compo Bio. Nut. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Opiates 392 Opioid receptors 84 Optimization 1 Opuntia ficus-indica 20 Oral contraceptive 201, 411 Oral hypoglycemic agents (OHA) 220 Oreochromis mossambicus 455 Organophosphate 451 Oridonin 59 Orthosiphon stamineus 417 Osteoblastic activity 363 Osteoblasts 254, 256 Osteogenic effect 355 Osteoporosis 364 Ostreolysin 251 Oxazepam 391 Oxidation 389 Oxidative DNA damage 275 Oxidative stress 431 Oxyacanthine 73 Oxyradicals 190, 202 Oyster Mushroom 251
p Paclitaxel 4, 5, 16, 379 Paepalanthus bromelioides 266 Paepalantin 266 Palmatine 73 Palmitate 234 Palmitoyl transferase activity 361 Panax ginseng 20, 54, 56 P. quinquefolius 194 Papaver somniferum 4 Papillomas 102 Papopmaceae 452 Paracetamol 391 Paralysis 71, 83, 224 Parasitemia 302 Parasiticide 231 Passiflora alata 212 P. incarnata 212 Pathogenesis 32, 68 Pathophysiology 17 Paullinia cupana 194 P-benzoquinone 69 P-benzoquinone-induced writhing reflex 75 P-benzoquinone-induced writhing test 72 PCR 435
PC-SPES 56 Penicillin 2, 4 Penicillium citrinum 5 P. notatum 2, 4 Pentobarbital 390 Pentobarbitone 391 Pentostatin 5 Peptic ulcer 408 Periodontitis 34, 87 Peritonitis 381 Peroxynitrous acid 163 Peroxysemiketals 212 Pesticidal 294 PGH 147 Pharmaceutical 68 Pharmacodynamics 17 Pharmacognosists 182 Pharmacognosy 48, 221 Pharmacokinetics 17,49,308,331,355, 399, 421 Pharmacological 355 Pharmacologists 182 Pharyngitis 34 Phellodendron sp. 74 Phenacetin 391 Phenobarbitone 337,339 Phenolic acids 357, 418 Phenylbutazone 365 Phenylephrine 361 Phenylpropanoid esters 213 Phenytoin 392, 395 Phloretin tangeretin 381 Phloridzin 368 Phorate 451 Phorbol esters 449 Phosphatidylinositol 3-kinase 440 Phospholipase-A2 enzyme 79 Phospholipids 362 Phosphorylation 442 Photooxidation 235 Phototoxicity 373 Physa fontinalis 451 P. occidentalis 456 Physiological pathways 46 Phytochemicals 45, 190, 226, 231, 266, 294 Phytoconstituents 226 Phytoestrogens 366 Phytohaemagglutinin 123 Phytomedicines 48, 198, 213, 239
Index
Phytopharmacology 198 Phytotherapy 68, 69, 198 Phytotoxic 456 Pila globosa 450 Pinellia ternata 56 Pioglitazone 20 Piper methysticum 212 Piroxicam 267, 268 Piscicides 449 Pistacia vera 68 Placental growth factor (PGF) 47 Plant adaptogens 308 Plantago major 95 P. ovate 20 Plants 67 Plasma 417 Plasma alanine 342 Plasma antioxidant status 421 Plasma concentration 321 Plasma membrane 390 Plasma potassium concentration 258 Plasmodium berghei 293, 295, 297 P. falciparum 294 Platanoside 79 Platelet-activating factor 68 Platelet-derived endothelial cell growth factor 47 Platelets 275 Pleiotrophin (PTN) 47 Pleurotolysin A 252 Pleurotolysin B 253 Pleurotus eryngii 252 P. ostreatus 251 PMNLs 138 Podophyllotoxin 16 Podophylum 5 Polyacetylenes 212 Polyarthritis 282 Polycyclic hydrocarbons 393 Polydypsia 220 Polygonum cuspidatum 53, 54 P. tinctorium 33 Polymorphism 337 Polymorphonuclear 136 Polymorphs 136 Polyphagia 220 Polypharmacy 33 Polyphenolic acid 418 Polyphenolic compounds 430 Polyphenols 425, 430
489 Polysaccharide 425 Polyunsaturated fatty acids 235 Polyuria 220 Polyvinylpolypyrrolidone 269 Poncirus trifoliata 381 Pongamia pinnata 455 Ponicidin 59 Pore forming protein 251 Poria cocus 54 Porphyromonas gingivalis 376 Positron emission tomography 9, 18 Post-feverfew syndrome 192 Potency test 283 PPAR 22 P-R interval 259 Pramlintide 20 Pravastatin sodium 410 Preclinical studies 18, 51 Predatory fishes 449 Prednisolone 158, 391, 408 Prednisone 391 Premature birth 282 Primordia 252 Probit analysis 233 Procarcinogens 393 Prochlorperazine 201 Progesterone 122 Progestins 122 Proinflammatory cytokines 159 Prokaryotic 6 Proliferin 47 Prophylaxis 165 Propranolol 391 Prostaglandin biosynthesis 78 Prostaglandins 68, 134, 140, 142, 146, 147,274 Protection rate 281 Protein metabolism 219 Proteoglycan 83 Proteomics 9 Protoanemonin 96 Protoberberine 73 Prunella vulgaris 82 Pruritus 220 Pseudohypericin 240 Pseudomonas aeruginosa 252, 297, 376 P. fluorescens 4 Psoriasis 31, 164 Psychological therapy 211 Psychopathies 308
490
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Psychopharmacological therapy 308 PTP-lB 22 Pulmonary edema 381 Putrescence 83 Pylorus cramps 212 P. ligature 274 Pyrazole 339 Pyrexia 73
Q Qing Dai 33 QRS complexes 259 Quality control (QC) 419 Quercetin 360 Quercetin 3-o-~-galactopyranoside 82 Quercetin 3-o-~-glucopyranoside 82 Quercetin-3, 7-o-a-dirhamnoside 107 Quinidine 392 Quinine 4, 5, 294
R Rabdosia rubescens 54 Radiotherapy 48, 53 Radix auricularia 451 Random allocation 182 Randomized 182 Randomized controlled trials 180 Ranunculin 96 Rauwolfia serpentina 5 Reactive oxygen species 189, 275, 361, 425 Real-time PCR 431 439 Recombinant DNA technology 2 Reducing oxidative stress 425 Reduction 389 Regression analysis 124 Remifemin® 213 Renal ischemia 361 Repaglinide 19 Reserpine 5, 365 Resiniferotoxin 454 Resveratrol 54 Retro-orbital plexus 223 Reverse pharmacology 2, 22 Rheumatism 67,76, 213 Rheumatoid arthritis 136 Rhodamine 368 Rhodiola rosea 308 Rhodioloside 308
Rhododendron chrysanthum 76 R. ferrugineum 76 Rhododendron ponticum 76 Ribonucleoside/nucleotide synthesis 389 Rifampicin 4, 339, 392, 393 Ritonavir 392 RNI165 Robinin 368 ROI 165 Rosa canina 98 Rosavin 308 Rosenroot 308 Rosiglitazone 20 Rosmarinic acid 84, 417 Rosmarinus officinalis 418 RT-PCR assay 372 Rubus sanctus 98 Rumex abyssinicus 94 R. crisp us 94 R. nervosus 94 R. patientia 94 Rutin 360
s Sabinene 455 Safety 1, 33, 219, 281 Safety test 283 Sakuranetin 368 Salicylaldehyde 332 Salmonella enteritidis 376 S. typhi 376 S. typhimurium 122, 233, 234, 370 Salvia 84 S. halophila 84 S. leriifolia 84 S. miltiorrhiza 53, 84 S. tomentosa 96 S. virgata 84 Sanguinaria sp. 74 Saponins 70, 226, 400, 449 Sarkomycin 5 Sarotherodon mossambica 454 Saudi folk medicine 71 Scandenone 93 Scavenging activity 92 Schima argenta 453 Schistosomiasis 449,450 Schizophrenia 308 Sciatica 98 Scopolamine butyl bromide 408
Index Scrophularia auriculata 102 S. ningpoensis 53 Scutellaria baicalensis 53, 54 S. lateriflora 212 Sedative 34 Sehima argenta 452 Selectins 134, 167 Selective serotonin reuptake inhibitors (SSRIS) 211 Selenium 19 Semisynthesis 1 Sempre viva 265 Serotonin 70,94, 140, 142, 144, 246 Seseli andronakii 70 S. gummiferum 70 S. indicum 70 S. libanotis 70 S. peucedanoides 70 S. resinosum 70 S. tortuosum 70 Sesquiterpenes lactones 449 Shaoyao-gancao-tang 408 Shigella flexneri 376 Shimadzu/IR-408 spectrophotometer 232 SHR-5308 Sialic acid 362 Sideritis ozturkii 85 Signal transduction 253 Sildenafil 18 Silibin 54 Silica gel column chromatography 296 Silicium dioxide 196 Silybum marianum 54 Silymarin 54 Sinuato199 Sinus arrhythmia 259 Sinusitis 69, 82 Sirolimus 5 Sister chromatid exchanges 121, 124 Sitaglitpin 20 Sitosterol 295 Skeletal system 356 Sleep disorders 212 Slugs 450 Smooth muscle 356 Snail 449 Snakebites 213 Solamargines 5 Solanum marginatum 5
491 Songarosaponins 102 S-oxidation 393 Spasmolytic flavonoid glycosides 212 Sphingomyelin (sm) 252 Sphingosine-I-phosphate 254 Spodoptera frugiperda 294 Spondylitis 87 Spongothymidine 2 Spongouridine 2 Sprague-Dawley rats 361,362, 419 St. John's wort 194, 240 Stachys lavandulifolia 212 Staphylococcus aureus 233, 234, 268, 296, 376 Stasis 138 Statin 363 Stearate 234 Stereochemistry 300 Steroids 33 Sterols 256 Stevia rebaudiana 20 Stigmasterol 295 Still birth 282 Stimulant 69 Stomachache 69 Streptococcus faecalis 407 S. pyogenes 233,234 Streptomyces 5 S. antibioticus 5 S. erythrochromogenes 5 S. fradiae 4 S. grise us 4 S. hygroscopicus 5 S. kanamyceticus 4 S. lincolnensis 4 S. peucetius 5 S. plicatus 5 S. roseosporus 4 S. tsukubaensis 5 S. venezuelae 4 S. vertic ill us 5 Streptomycin 4, 389, 390 Stress 32 Strobilanthes flaccidifolius 33 Structure elucidation 1 Students t-test 124, 298, 422 Subcellular fractions 335 Succinylcholine 397 Suicide-commandos 135 Sulfasalazine 391
492
Compo Bio. Nat. Prod., Vol. 2 - Efficacy, Safety & Clinical Evaluation I
Sulfate 389 Sulfate conjugation 395 Sulfonylureas 220 Supercritical fluid 9 Supercritical fluid chromatography (SFC) 9 Superoxide anion radical 189 Superoxide anions 374 Superoxide scavengers 374 Surrogate markers 60 Swiss albino mice 214, 222, 267, 297 Symptomatic relief 68 Synergistic effect 221 Synthetic compounds 449 Synthetic drugs 408 Synthetic pyrethroids 451 Syrian hamster 334
T Tacrolimus 5 Tail suspension test 239 Tamus communis 76 Tanacetum parthenium 192, 194 Tangeretin 367 Tanin 452 Tannins 225, 449 Taxifolin 381 Taxus brevifolia 5, 54 T. chinensis 53 Taxusin 106 Teicoplanin 4 Tenderness 39 Terpenoids 449 Tethya crypta 2 Tetramers 254 TGF-b 58 Thea olesosa 452, 453 Therapeutic 310, 406 Therapeutic agents 2, 41, 221 Therapeutic potential 31, 68 Thermodynamics 9 Thermoregulation 144 Thiazolidinediones 220 Thiobarbituric acid (TBA) 371, 418 Thiobarbituric acid reactive substances (TBARS) 421 Thiopental 390 Thiouracil 396 Threo-trans-threo-trans-threo 300 Thrombin 410
Thrombospondin 47 Thromboxane a2 147 Thyroid peroxidase (TPO) 365 Tilia argentea 107 Tiliroside 79, 108 Tinyatoxin 454 T -kininogen 146 Tmax 406 TNF-a 79, 106, 150,372,431 TNF-13 58 Tobacco 313 Tocopherols 368 Tolazamide 19 Tolbutamide 19 Tolypocladium inflatum 5 Tonsillitis 34 Topotecan 16 Toxic 449 Toxicity 224, 251, 252, 303, 331, 362 Toxins 356 Toxylon pomiferum 92 Traditional chinese herbal medicine 3 Traditional chinese medicine (TCM) 31, 53, 190 Transforming growth factor-a (TGF-A) 47 Transforming growth factor-13 (TGF-b) 47 Translational research 2 Tranylcypromine 201 Trasina 221 Trauma 32, 83 Trematodes 450 Tricarboxylic acid cycle 220 Trichlorofon 451 Trichlroacetic acid 418 Trichosanthes kirilowii 82 Trifluorperazine 201 Triglycerides 431 Triglycerols 360 Trigonella foenum-graecum 19,20 Trihydroxyethylrutin 373 Tristetraprolin 430 Triterpene sterols 400 Triterpenes 225 Trolox 422 Tryptanthrin 40 Tukey's multiple range tests 214 Tumor necrosis 76 Tumor necrosis factor (TNF) 369, 430
493
Index
Tumour 356 Tumour endothelial cells (TEe) 50 Tumour necrosis factor-a (TNF -a) 47 Tumour xenografts 50 Turkey 67 Turkish folk medicine 71 Turnera aphrodisiaca 212 Turpentine 144 T-wave 259 Tyrosol308
u Uapaca guinensis 453 Ulcerative lesion index (ULI) 267 illcerogenicity 71 Urinary infections 69 Ursolic 77 Ursolic acid 77,79 Uteritis 282 UV spectra 10 UV spectrum 316 Uvaria hookeri 235 U. narum 235
v Vacuolation 342 Vagotomy 274 Valeriana eduli 212 V. officinalis 212 V. thalictroides 212 V. walliichii 212 Vanadium 19 Vancomycin 4 Varian cp-3380 gas chromatograph 232 Vascular endothelial growth factor (VEGF) 47 Vascular permeability factor (VPF) 47 Vasoconstrictor 78, 148 Vasodilation 138 Vasorelaxant effect 364, 361
Vasospasm 261 VEGF 46 VEGFA-B 435 Verapamil 379, 392 Verbascosaponin a 100 Verbascoside 86 Verbascum lasianthum 99 V. pterocalycinum 102 V. songaricum 102 Verminoside 103 Veronica anagallis-aquatica 103 Verproside 103 Vinblastin 5 Vinca rosea 5 Vincristine 4 Viscum album 87 Vitis vinifera 109 Vivarium 242 Voglibose 5
w Warfarin 392 Wistar rats 310, 362, 432 World health organization 6
x Xanthomicrol 86 Xanthophyllum sp. 74 Xanthotoxin 456 Xenobiotic 389 Xenoestrogens 366 Xhantones 265 Xylopia aromatica 294
z Zingiber officinale 54 Zinophos 451 Zizyphus jujuba 56 Zoonotic 282